Academic Appointments


Honors & Awards


  • Prostate Cancer Research Award, CaP CURE (1993,1995,1996,1997,1999,2000)
  • Prostate Cancer Research Award, Prostate Cancer Foundation (2003, 2008)
  • Cardura International Competitive Award, Pfizer (2000/2001)

Professional Education


  • B.S., Stanford University, Biology (1974)
  • Ph.D., University of Colorado, biology (1979)

Current Research and Scholarly Interests


The overall goal of my research is to develop realistic experimental models of benign and malignant prostatic diseases. Both benign prostatic hyperplasia (BPH) and prostate cancer (PCa) are major medical problems, causing significant morbidity and mortality. My lab has developed techniques to establish primary cultures of epithelial and stromal cells from normal, benign or malignant human prostatic tissues, and we have used these cell cultures to investigate many aspects of the molecular and cellular biology of the prostate. More recently, we have established “tissue slice cultures” (TSCs), which are live, precision-cut thin sections of tissues that can be maintained in culture for several days. TSCs are perhaps the most representative experimental model of the human prostate available, containing almost all of the cells typically present in the body and maintaining essential epithelial-stromal interactions as well as differentiated cells, which are typically lost in mono-culture. TSCs can also be established as grafts under the renal capsule of mice, providing an in vivo model of the benign and malignant prostate. In addition to our cell and tissue models, our research takes advantage of the archival patient materials available in the Department of Urology. These include tissues from a well-characterized series of radical prostatectomy specimens and a serum bank. Using these diverse models and resources, we carry out studies related to diagnosis, prognosis and treatment of PCa. Currently, some of our projects include: 1) investigating the association of a splice-variant of the androgen receptor with aggressive PCa, 2) studying cell surface proteoglycans as novel and specific biomarkers of PCa, 3) searching for serum autoantibodies or proteins that could be used to diagnose PCa, 4) creating induced pluripotent stem (iPS) cells from PCa cells as a novel model to characterize cancer-related methylation, 5) determining the role of monoamine oxidase A (MAOA) in normal prostate differentiation and as a therapeutic target, 6) using TSCs to screen phage display libraries to discover PCa-specific cell surface molecules for cancer-specific targeting, 7) developing hyperpolarized 13C-pyruvate magnetic resonance spectroscopic imaging for rapid evaluation of therapeutic response, 8) creating additional cell culture models of metastatic PCa, and 9) testing the efficacy of a novel organic arsenical compound against PCa. These projects are funded in part by the NIH, the DoD, and the Prostate Cancer Foundation.

2022-23 Courses


Graduate and Fellowship Programs


All Publications


  • Animal Models and Their Role in Imaging-Assisted Co-Clinical Trials. Tomography (Ann Arbor, Mich.) Peehl, D. M., Badea, C. T., Chenevert, T. L., Daldrup-Link, H. E., Ding, L., Dobrolecki, L. E., Houghton, A. M., Kinahan, P. E., Kurhanewicz, J., Lewis, M. T., Li, S., Luker, G. D., Ma, C. X., Manning, H. C., Mowery, Y. M., O'Dwyer, P. J., Pautler, R. G., Rosen, M. A., Roudi, R., Ross, B. D., Shoghi, K. I., Sriram, R., Talpaz, M., Wahl, R. L., Zhou, R. 2023; 9 (2): 657-680

    Abstract

    The availability of high-fidelity animal models for oncology research has grown enormously in recent years, enabling preclinical studies relevant to prevention, diagnosis, and treatment of cancer to be undertaken. This has led to increased opportunities to conduct co-clinical trials, which are studies on patients that are carried out parallel to or sequentially with animal models of cancer that mirror the biology of the patients' tumors. Patient-derived xenografts (PDX) and genetically engineered mouse models (GEMM) are considered to be the models that best represent human disease and have high translational value. Notably, one element of co-clinical trials that still needs significant optimization is quantitative imaging. The National Cancer Institute has organized a Co-Clinical Imaging Resource Program (CIRP) network to establish best practices for co-clinical imaging and to optimize translational quantitative imaging methodologies. This overview describes the ten co-clinical trials of investigators from eleven institutions who are currently supported by the CIRP initiative and are members of the Animal Models and Co-clinical Trials (AMCT) Working Group. Each team describes their corresponding clinical trial, type of cancer targeted, rationale for choice of animal models, therapy, and imaging modalities. The strengths and weaknesses of the co-clinical trial design and the challenges encountered are considered. The rich research resources generated by the members of the AMCT Working Group will benefit the broad research community and improve the quality and translational impact of imaging in co-clinical trials.

    View details for DOI 10.3390/tomography9020053

    View details for PubMedID 36961012

    View details for PubMedCentralID PMC10037611

  • Multiparametric Magnetic Resonance Imaging and Metabolic Characterization of Patient-Derived Xenograft Models of Clear Cell Renal Cell Carcinoma. Metabolites Agudelo, J. P., Upadhyay, D., Zhang, D., Zhao, H., Nolley, R., Sun, J., Agarwal, S., Bok, R. A., Vigneron, D. B., Brooks, J. D., Kurhanewicz, J., Peehl, D. M., Sriram, R. 2022; 12 (11)

    Abstract

    Patient-derived xenografts (PDX) are high-fidelity cancer models typically credentialled by genomics, transcriptomics and proteomics. Characterization of metabolic reprogramming, a hallmark of cancer, is less frequent. Dysregulated metabolism is a key feature of clear cell renal cell carcinoma (ccRCC) and authentic preclinical models are needed to evaluate novel imaging and therapeutic approaches targeting metabolism. We characterized 5 PDX from high-grade or metastatic ccRCC by multiparametric magnetic resonance imaging (MRI) and steady state metabolic profiling and flux analysis. Similar to MRI of clinical ccRCC, T2-weighted images of orthotopic tumors of most PDX were homogeneous. The increased hyperintense (cystic) areas observed in one PDX mimicked the cystic phenotype typical of some RCC. The negligible hypointense (necrotic) areas of PDX grown under the highly vascularized renal capsule are beneficial for preclinical studies. Mean apparent diffusion coefficient (ADC) values were equivalent to those of ccRCC in human patients. Hyperpolarized (HP) [1-13C]pyruvate MRI of PDX showed high glycolytic activity typical of high-grade primary and metastatic ccRCC with considerable intra- and inter-tumoral variability, as has been observed in clinical HP MRI of ccRCC. Comparison of steady state metabolite concentrations and metabolic flux in [U-13C]glucose-labeled tumors highlighted the distinctive phenotypes of two PDX with elevated levels of numerous metabolites and increased fractional enrichment of lactate and/or glutamate, capturing the metabolic heterogeneity of glycolysis and the TCA cycle in clinical ccRCC. Culturing PDX cells and reimplanting to generate xenografts (XEN), or passaging PDX in vivo, altered some imaging and metabolic characteristics while transcription remained like that of the original PDX. These findings show that PDX are realistic models of ccRCC for imaging and metabolic studies but that the plasticity of metabolism must be considered when manipulating PDX for preclinical studies.

    View details for DOI 10.3390/metabo12111117

    View details for PubMedID 36422257

  • The controversial role and therapeutic development of the m6A demethylase FTO in renal cell carcinoma. Translational oncology Zhang, D., Wornow, S., Peehl, D. M., Rankin, E. B., Brooks, J. D. 2022; 25: 101518

    Abstract

    Fat mass and obesity-associated (FTO) protein, the first m6A demethylase identified in 2011, regulates multiple aspects of RNA biology including splicing, localization, stability, and translation. Accumulating data show that FTO is involved in numerous physiological processes and is implicated in multiple cancers including renal cell carcinoma (RCC). However, the exact role of FTO in RCC remains controversial. Some studies demonstrated that decreased FTO expression was associated with aggressive clinical features and shorter overall survival in clear cell RCC (ccRCC) patients, while others found that FTO inhibition selectively reduced the growth and survival of VHL-deficient ccRCC cells in vitro and in vivo. Here, we review the evidence supporting either a promoting or suppressive role of FTO in kidney cancers, the mechanisms of action of FTO, and recent progress in developing FTO inhibitors.

    View details for DOI 10.1016/j.tranon.2022.101518

    View details for PubMedID 36037557

  • SU086, an inhibitor of HSP90, impairs glycolysis and represents a treatment strategy for advanced prostate cancer. Cell reports. Medicine Rice, M. A., Kumar, V., Tailor, D., Garcia-Marques, F. J., Hsu, E., Liu, S., Bermudez, A., Kanchustambham, V., Shankar, V., Inde, Z., Alabi, B. R., Muruganantham, A., Shen, M., Pandrala, M., Nolley, R., Aslan, M., Ghoochani, A., Agarwal, A., Buckup, M., Kumar, M., Going, C. C., Peehl, D. M., Dixon, S. J., Zare, R. N., Brooks, J. D., Pitteri, S. J., Malhotra, S. V., Stoyanova, T. 2022; 3 (2): 100502

    Abstract

    Among men, prostate cancer is the second leading cause of cancer-associated mortality, with advanced disease remaining a major clinical challenge. We describe a small molecule, SU086, as a therapeutic strategy for advanced prostate cancer. We demonstrate that SU086 inhibits the growth of prostate cancer cells invitro, cell-line and patient-derived xenografts invivo, and exvivo prostate cancer patient specimens. Furthermore, SU086 in combination with standard of care second-generation anti-androgen therapies displays increased impairment of prostate cancer cell and tumor growth invitro and invivo. Cellular thermal shift assay reveals that SU086 binds to heat shock protein 90 (HSP90) and leads to a decrease in HSP90 levels. Proteomic profiling demonstrates that SU086 binds to and decreases HSP90. Metabolomic profiling reveals that SU086 leads to perturbation of glycolysis. Our study identifies SU086 as a treatment for advanced prostate cancer as a single agent or when combined with second-generation anti-androgens.

    View details for DOI 10.1016/j.xcrm.2021.100502

    View details for PubMedID 35243415

  • Identifying a novel glycolytic inhibitor for treatment of aggressive prostate cancer. Stoyanova, T., Rice, M. A., Kumar, V., Tailor, D., Garcia-Marques, F., Bermudez, A., Kanchustambham, V., Shankar, V., Inde, Z., Pandrala, M., Nolley, R., Ghoochani, A., Liu, S., Aslan, M., Agarwal, A., Buckup, M., Hsu, E., Going, C. C., Peehl, D. M., Dixon, S. J., Zare, R. N., Brooks, J. D., Pitteri, S. J., Malhotra, S. V., Stoyanova, T. AMER ASSOC CANCER RESEARCH. 2021
  • MAP3K7 loss drives enhanced androgen signaling and independently confers risk of recurrence in prostate cancer with joint loss of CHD1. Molecular cancer research : MCR Jillson, L. K., Rider, L. C., Rodrigues, L. U., Romero, L., Karimpour-Fard, A., Nieto, C., Gillette, C., Torkko, K., Danis, E., Smith, E. E., Nolley, R., Peehl, D. M., Lucia, M. S., Costello, J. C., Cramer, S. D. 2021

    Abstract

    Prostate cancer (PCa) genomic subtypes that stratify aggressive disease and inform treatment decisions at the primary stage are currently limited. Previously, we functionally validated an aggressive subtype present in 15% of PCa characterized by dual deletion of MAP3K7 and CHD1. Recent studies in the field have focused on deletion of CHD1 and its role in androgen receptor (AR) chromatin distribution and resistance to AR-targeted therapy, however, CHD1 is rarely lost without co-deletion of MAP3K7. Here we show that in the clinically relevant context of co-loss of MAP3K7 and CHD1 there are significant, collective changes to aspects of AR signaling. While CHD1 loss mainly impacts the expansion of the AR cistrome, loss of MAP3K7 drives increased AR target gene expression. PCa cell line models engineered to co-suppress MAP3K7 and CHD1 also demonstrated increased AR-v7 expression and resistance to the AR-targeting drug enzalutamide. Furthermore, we determined that low protein expression of both genes is significantly associated with biochemical recurrence (BCR) in a clinical cohort of radical prostatectomy specimens. Low MAP3K7 expression, however, was the strongest independent predictor for risk of BCR over all other tested clinicopathologic factors including CHD1 expression. Collectively, these findings illustrate the importance of MAP3K7 loss in a molecular subtype of PCa that poses challenges to conventional therapeutic approaches. Implications: These findings strongly implicate MAP3K7 loss as a biomarker for aggressive prostate cancer with significant risk for recurrence that poses challenges for conventional androgen receptor-targeted therapies.

    View details for DOI 10.1158/1541-7786.MCR-20-0913

    View details for PubMedID 33846123

  • Trop2 is a driver of metastatic prostate cancer with neuroendocrine phenotype via PARP1. Proceedings of the National Academy of Sciences of the United States of America Hsu, E. C., Rice, M. A., Bermudez, A. n., Marques, F. J., Aslan, M. n., Liu, S. n., Ghoochani, A. n., Zhang, C. A., Chen, Y. S., Zlitni, A. n., Kumar, S. n., Nolley, R. n., Habte, F. n., Shen, M. n., Koul, K. n., Peehl, D. M., Zoubeidi, A. n., Gambhir, S. S., Kunder, C. A., Pitteri, S. J., Brooks, J. D., Stoyanova, T. n. 2020

    Abstract

    Resistance to androgen deprivation therapy, or castration-resistant prostate cancer (CRPC), is often accompanied by metastasis and is currently the ultimate cause of prostate cancer-associated deaths in men. Recently, secondary hormonal therapies have led to an increase of neuroendocrine prostate cancer (NEPC), a highly aggressive variant of CRPC. Here, we identify that high levels of cell surface receptor Trop2 are predictive of recurrence of localized prostate cancer. Moreover, Trop2 is significantly elevated in CRPC and NEPC, drives prostate cancer growth, and induces neuroendocrine phenotype. Overexpression of Trop2 induces tumor growth and metastasis while loss of Trop2 suppresses these abilities in vivo. Trop2-driven NEPC displays a significant up-regulation of PARP1, and PARP inhibitors significantly delay tumor growth and metastatic colonization and reverse neuroendocrine features in Trop2-driven NEPC. Our findings establish Trop2 as a driver and therapeutic target for metastatic prostate cancer with neuroendocrine phenotype and suggest that high Trop2 levels could identify cancers that are sensitive to Trop2-targeting therapies and PARP1 inhibition.

    View details for DOI 10.1073/pnas.1905384117

    View details for PubMedID 31932422

  • Elevated Tumor Lactate and Efflux in High-grade Prostate Cancer demonstrated by Hyperpolarized 13C Magnetic Resonance Spectroscopy of Prostate Tissue Slice Cultures. Cancers Sriram, R. n., Van Criekinge, M. n., DeLos Santos, J. n., Ahamed, F. n., Qin, H. n., Nolley, R. n., Santos, R. D., Tabatabai, Z. L., Bok, R. A., Keshari, K. R., Vigneron, D. B., Peehl, D. M., Kurhanewicz, J. n. 2020; 12 (3)

    Abstract

    Non-invasive assessment of the biological aggressiveness of prostate cancer (PCa) is needed for men with localized disease. Hyperpolarized (HP) 13C magnetic resonance (MR) spectroscopy is a powerful approach to image metabolism, specifically the conversion of HP [1-13C]pyruvate to [1-13C]lactate, catalyzed by lactate dehydrogenase (LDH). Significant increase in tumor lactate was measured in high-grade PCa relative to benign and low-grade cancer, suggesting that HP 13C MR could distinguish low-risk (Gleason score ≤3 + 4) from high-risk (Gleason score ≥4 + 3) PCa. To test this and the ability of HP 13C MR to detect these metabolic changes, we cultured prostate tissues in an MR-compatible bioreactor under continuous perfusion. 31P spectra demonstrated good viability and dynamic HP 13C-pyruvate MR demonstrated that high-grade PCa had significantly increased lactate efflux compared to low-grade PCa and benign prostate tissue. These metabolic differences are attributed to significantly increased LDHA expression and LDH activity, as well as significantly increased monocarboxylate transporter 4 (MCT4) expression in high- versus low- grade PCa. Moreover, lactate efflux, LDH activity, and MCT4 expression were not different between low-grade PCa and benign prostate tissues, indicating that these metabolic alterations are specific for high-grade disease. These distinctive metabolic alterations can be used to differentiate high-grade PCa from low-grade PCa and benign prostate tissues using clinically translatable HP [1-13C]pyruvate MR.

    View details for DOI 10.3390/cancers12030537

    View details for PubMedID 32110965

  • The m6A RNA demethylase FTO is a HIF-independent synthetic lethal partner with the VHL tumor suppressor. Proceedings of the National Academy of Sciences of the United States of America Xiao, Y. n., Thakkar, K. N., Zhao, H. n., Broughton, J. n., Li, Y. n., Seoane, J. A., Diep, A. N., Metzner, T. J., von Eyben, R. n., Dill, D. L., Brooks, J. D., Curtis, C. n., Leppert, J. T., Ye, J. n., Peehl, D. M., Giaccia, A. J., Sinha, S. n., Rankin, E. B. 2020

    Abstract

    Loss of the von Hippel-Lindau (VHL) tumor suppressor is a hallmark feature of renal clear cell carcinoma. VHL inactivation results in the constitutive activation of the hypoxia-inducible factors (HIFs) HIF-1 and HIF-2 and their downstream targets, including the proangiogenic factors VEGF and PDGF. However, antiangiogenic agents and HIF-2 inhibitors have limited efficacy in cancer therapy due to the development of resistance. Here we employed an innovative computational platform, Mining of Synthetic Lethals (MiSL), to identify synthetic lethal interactions with the loss of VHL through analysis of primary tumor genomic and transcriptomic data. Using this approach, we identified a synthetic lethal interaction between VHL and the m6A RNA demethylase FTO in renal cell carcinoma. MiSL identified FTO as a synthetic lethal partner of VHL because deletions of FTO are mutually exclusive with VHL loss in pan cancer datasets. Moreover, FTO expression is increased in VHL-deficient ccRCC tumors compared to normal adjacent tissue. Genetic inactivation of FTO using multiple orthogonal approaches revealed that FTO inhibition selectively reduces the growth and survival of VHL-deficient cells in vitro and in vivo. Notably, FTO inhibition reduced the survival of both HIF wild type and HIF-deficient tumors, identifying FTO as an HIF-independent vulnerability of VHL-deficient cancers. Integrated analysis of transcriptome-wide m6A-seq and mRNA-seq analysis identified the glutamine transporter SLC1A5 as an FTO target that promotes metabolic reprogramming and survival of VHL-deficient ccRCC cells. These findings identify FTO as a potential HIF-independent therapeutic target for the treatment of VHL-deficient renal cell carcinoma.

    View details for DOI 10.1073/pnas.2000516117

    View details for PubMedID 32817424

  • S100A10 is a critical mediator of GAS6/AXL-induced angiogenesis in renal cell carcinoma. Cancer research Xiao, Y., Zhao, H., Tian, L., Nolley, R., Diep, A. N., Ernst, A., Fuh, K. C., Miao, Y. R., von Eyben, R., Leppert, J. T., Brooks, J. D., Peehl, D. M., Giaccia, A. J., Rankin, E. B. 2019

    Abstract

    Angiogenesis is a hallmark of cancer that promotes tumor progression and metastasis. However, antiangiogenic agents have limited efficacy in cancer therapy due to the development of resistance. In clear cell renal cell carcinoma (ccRCC), AXL expression is associated with antiangiogenic resistance and poor survival. Here, we establish a role for GAS6/AXL signaling in promoting the angiogenic potential of ccRCC cells through the regulation of the plasminogen receptor S100A10. Genetic and therapeutic inhibition of AXL signaling in ccRCC tumor xenografts reduced tumor vessel density and growth under the renal capsule. GAS6/AXL signaling activated the expression of S100A10 through SRC to promote plasmin production, endothelial cell invasion and angiogenesis. Importantly, treatment with the small molecule AXL inhibitor cabozantinib or an ultra-high affinity soluble AXL Fc fusion decoy receptor (sAXL) reduced the growth of a pazopanib-resistant ccRCC patient-derived xenograft. Moreover, the combination of sAXL synergized with pazopanib and axitinib to reduce ccRCC patient-derived xenograft growth and vessel density. These findings highlight a role for AXL/S100A10 signaling in mediating the angiogenic potential of ccRCC cells and support the combination of AXL inhibitors with antiangiogenic agents for advanced ccRCC.

    View details for DOI 10.1158/0008-5472.CAN-19-1366

    View details for PubMedID 31585940

  • miR-22 Regulates Invasion, Gene Expression and Predicts Overall Survival in Patients with Clear Cell Renal Cell Carcinoma. Kidney cancer Gong, X., Zhao, H., Saar, M., Peehl, D. M., Brooks, J. D. 2019; 3 (2): 119–32

    Abstract

    Background: Clear cell renal cell carcinoma (ccRCC) is molecularly diverse and distinct molecular subtypes show different clinical outcomes. MicroRNAs (miRNAs) are essential components of gene regulatory networks and play a crucial role in progression of many cancer types including ccRCC.Objective: Identify prognostic miRNAs and determine the role of miR-22 in ccRCC.Methods: Hierarchical clustering was done in R using gene expression profiles of over 450 ccRCC cases in The Cancer Genome Atlas (TCGA). Kaplan-Meier analysis was performed to identify prognostic miRNAs in the TCGA dataset. RNA-Seq was performed to identify miR-22 target genes in primary ccRCC cells and Matrigel invasion assay was performed to assess the effects of miR-22 overexpression on cell invasion.Results: Hierarchical clustering analysis using 2,621 prognostic genes previously identified by our group demonstrated that ccRCC patients with longer overall survival expressed lower levels of genes promoting proliferation or immune responses, while better maintaining gene expression associated with cortical differentiation and cell adhesion. Targets of 26 miRNAs were significantly enriched in the 2,621 prognostic genes and these miRNAs were prognostic by themselves. MiR-22 was associated with poor overall survival in the TCGA dataset. Overexpression of miR-22 promoted invasion of primary ccRCC cells in vitro and modulated transcriptional programs implicated in cancer progression including DNA repair, cell proliferation and invasion.Conclusions: Our results suggest that ccRCCs with differential clinical outcomes have distinct transcriptomes for which miRNAs could serve as master regulators. MiR-22, as a master regulator, promotes ccRCC progression at least in part by enhancing cell invasion.

    View details for DOI 10.3233/KCA-190051

    View details for PubMedID 31763513

  • Defining new drivers of castration- resistant prostate cancer Hsu, E., Rice, M., Nolley, R., Bermudez, A., Huang, J., Peehl, D., Kunder, C., Pitteri, S., Brooks, J., Stoyanova, T. AMER ASSOC CANCER RESEARCH. 2018: 90
  • A note on improved statistical approaches to account for pseudoprogression CANCER CHEMOTHERAPY AND PHARMACOLOGY Abrouk, N., Oronsky, B., Caroen, S., Ning, S., Knox, S., Peehl, D. 2018; 81 (3): 621–26

    Abstract

    Responses to immuno-oncology agents are often subject to misinterpretation as apparent tumor growth due to immune infiltration leads to the appearance of progressive disease and can result in the discontinuation of effective therapeutic agents. Better statistical strategies to determine experimental outcomes are needed to distinguish between true and pseudoprogression. We applied time-to-event statistical analyses methods that account for study design features and capture the longitudinal and panoramic aspects of pseudoprogression to test superiority of a combination of RRx-001, a novel tumor-associated macrophage polarizing agent in Phase 2, and an anti-PD-L1 antibody in a myeloma preclinical model, comparing to traditional, mean-based mixed effects modeling approaches that did not show statistical significance. Nonparametric p values for the difference of cumulative incidence rates of time to ≥ 50% tumor growth reduction and its associated restricted mean survival times are computed and found to be statistically significant. Kaplan-Meier description of time-to-volume reduction (≥ 50%) coupled with Cox's proportional hazards model follows the data longitudinally and therefore permits an analysis of immune infiltration resolution, making it an improved method for analysis of preclinical experiments with immuno-oncology agents.

    View details for PubMedID 29404682

  • Sensitization of neuroendocrine prostate cancer by RRx-001. Peehl, D., Zhao, H., Ning, S. AMER SOC CLINICAL ONCOLOGY. 2018
  • A spliced form of CD44 expresses the unique glycan that is recognized by the prostate cancer specific antibody F77 ONCOTARGET Chen, X., Nagai, Y., Zhu, Z., Ruan, H., Peehl, D. M., Greene, M. I., Zhang, H. 2018; 9 (3): 3631–40

    Abstract

    Prostate cancer is the most common cancer occurring in men in the United States. The monoclonal antibody F77 that was originally developed in our laboratory recognizes mainly glycolipids as well as O-linked glycosylation on proteins in prostate cancer cells. We have identified a spliced form of glycoprotein CD44 as one critical protein expressing the F77 antigen. The F77-specific glycosylation occurs on multiple potential glycosylation sites on the CD44 protein encoded by the fourteenth exon. CD44 is a tumor stem cell marker and is known to induce tumor stemness and metastasis. Knockdown of CD44 or FUT1 genes dramatically reduced F77-induced apoptosis in prostate cancer cell lines. We developed an ELISA using both a CD44 antibody and F77 to identify the special form of glycosylated CD44 from prostate cancer cells as well as from serum samples of prostate cancer patients. These results reveal a CD44-dependent mechanism for F77 to induce tumor cell apoptosis, and a new strategy for the detection of glycosylated CD44 proteins secreted by prostate cancer cells.

    View details for PubMedID 29423071

  • Bioorthogonal Labeling of Human Prostate Cancer Tissue Slice Cultures for Glycoproteomics ANGEWANDTE CHEMIE-INTERNATIONAL EDITION Spiciarich, D. R., Nolley, R., Maund, S. L., Purcell, S. C., Herschel, J., Iavarone, A. T., Peehl, D. M., Bertozzi, C. R. 2017; 56 (31): 8992–97

    Abstract

    Sialylated glycans are found at elevated levels in many types of cancer and have been implicated in disease progression. However, the specific glycoproteins that contribute to the cancer cell-surface sialylation are not well characterized, specifically in bona fide human disease tissue. Metabolic and bioorthogonal labeling methods have previously enabled the enrichment and identification of sialoglycoproteins from cultured cells and model organisms. Herein, we report the first application of this glycoproteomic platform to human tissues cultured ex vivo. Both normal and cancerous prostate tissues were sliced and cultured in the presence of the azide-functionalized sialic acid biosynthetic precursor Ac4 ManNAz. The compound was metabolized to the azidosialic acid and incorporated into cell surface and secreted sialoglycoproteins. Chemical biotinylation followed by enrichment and mass spectrometry led to the identification of glycoproteins that were found at elevated levels or uniquely in cancerous prostate tissue. This work therefore extends the use of bioorthogonal labeling strategies to problems of clinical relevance.

    View details for PubMedID 28649697

    View details for PubMedCentralID PMC5675001

  • Comprehensive Drug Testing of Patient-derived Conditionally Reprogrammed Cells from Castration-resistant Prostate Cancer EUROPEAN UROLOGY Saeed, K., Rahkama, V., Eldfors, S., Bychkov, D., Mpindi, J. P., Yadav, B., Paavolainen, L., Aittokallio, T., Heckman, C., Wennerberg, K., Peehl, D. M., Horvath, P., Mirtti, T., Rannikko, A., Kallioniemi, O., Ostling, P., af Hallstrom, T. M. 2017; 71 (3): 319-327

    Abstract

    Technology development to enable the culture of human prostate cancer (PCa) progenitor cells is required for the identification of new, potentially curative therapies for PCa.We established and characterized patient-derived conditionally reprogrammed cells (CRCs) to assess their biological properties and to apply these to test the efficacies of drugs.CRCs were established from seven patient samples with disease ranging from primary PCa to advanced castration-resistant PCa (CRPC). The CRCs were characterized by genomic, transcriptomic, protein expression, and drug profiling.The phenotypic quantification of the CRCs was done based on immunostaining followed by image analysis with Advanced Cell Classifier using Random Forest supervised machine learning. Copy number aberrations (CNAs) were called from whole-exome sequencing and transcriptomics using in-house pipelines. Dose-response measurements were used to generate multiparameter drug sensitivity scores using R-statistical language.We generated six benign CRC cultures which all had an androgen receptor-negative, basal/transit-amplifying phenotype with few CNAs. In three-dimensional cell culture, these cells could re-express the androgen receptor. The CRCs from a CRPC patient (HUB.5) displayed multiple CNAs, many of which were shared with the parental tumor. We carried out high-throughput drug-response studies with 306 emerging and clinical cancer drugs. Using the benign CRCs as controls, we identified the Bcl-2 family inhibitor navitoclax as the most potent cancer-specific drug for the CRCs from a CRPC patient. Other drug efficacies included taxanes, mepacrine, and retinoids.Comprehensive cancer pharmacopeia-wide drug testing of CRCs from a CRPC patient highlighted both known and novel drug sensitivities in PCa, including navitoclax, which is currently being tested in clinical trials of CRPC.We describe an approach to generate patient-derived cancer cells from advanced prostate cancer and apply such cells to discover drugs that could be applied in clinical trials for castration-resistant prostate cancer.

    View details for DOI 10.1016/j.eururo.2016.04.019

    View details for Web of Science ID 000396333700008

    View details for PubMedID 27160946

  • The immunomodulatory anticancer agent, RRx-001, induces an interferon response through epigenetic induction of viral mimicry CLINICAL EPIGENETICS Zhao, H., Ning, S., Nolley, R., Scicinski, J., Oronsky, B., Knox, S. J., Peehl, D. M. 2017; 9

    Abstract

    RRx-001, a dinitroazetidine derivative, is a novel anticancer agent currently in phase II clinical trials. It mediates immunomodulatory effects either directly through polarization of tumor associated macrophages or indirectly through vascular normalization and increased T-lymphocyte infiltration. With multiple additional mechanisms of action including upregulation of oxidative stress, depletion of GSH and NADPH, anti-angiogenesis and epigenetic modulation, RRx-001 is being studied as a radio- and chemo-sensitizer to resensitize tumors to prior therapy and to prime tumors to respond to radiation, chemotherapy and immunotherapy in combination therapy studies. Here, we identified another mechanism, viral mimicry, which refers to the "unsilencing" of epigenetically repressed viral genes present in the tumor that provokes an immune response and may contribute to the anticancer activity of RRx-001.RRx-001 inhibited the growth of colon cancer cells (HCT 116) and decreased levels of the DNA methyltransferases DNMT1 and DNMT3a in a time and dose-dependent manner. Treatment of HCT 116 cells with 0.5 μM RRx-001 for 24 h significantly increased transcripts of interferon (IFN)-responsive genes and this induction was sustained for up to 4 weeks after transient exposure to RRx-001. ELISA assays showed that RRx-001 increased secretion of type I and III IFNs by HCT 116 cells, and these IFNs were confirmed to be bioactive. Transcription of endogenous retrovirus ERV-Fc2 and LTRs from the ERV-L family (MLT2B4 and MLT1C49) was induced by RRx-001. The induction of ERV-Fc2-env was through demethylation of ERV-Fc2 LTR as determined by methylation-specific polymerase chain reaction and combined bisulfite restriction analysis. Immunofluorescence staining with J2 antibody confirmed induction of double-stranded RNA.Transient exposure of HCT 116 cells to low-dose RRx-001 induced transcription of silenced retroviral genes present in the cancer cell DNA with subsequent synthesis of IFN in response to this "pseudo-pathogenic" stimulus, mimicking an antiviral defense. RRx-001-mediated IFN induction may have the potential to improve the efficacy of immunotherapies as well as radiotherapy, standard chemotherapies and molecularly targeted agents when used in combination. The striking safety profile of RRx-001 in comparison to other more toxic epigenetic and immunomodulatory agents such as azacitidine makes it a leading candidate for such clinical applications.

    View details for DOI 10.1186/s13148-017-0312-z

    View details for Web of Science ID 000392764400001

    View details for PubMedID 28149332

    View details for PubMedCentralID PMC5270305

  • RRx-001: a systemically non-toxic M2-to-M1 macrophage stimulating and prosensitizing agent in Phase II clinical trials EXPERT OPINION ON INVESTIGATIONAL DRUGS Oronsky, B., Paulmurugan, R., Foygel, K., Scicinski, J., Knox, S. J., Peehl, D., Zhao, H., Ning, S., Cabrales, P., Summers, T. A., Reid, T. R., Fitch, W. L., Kim, M. M., Trepel, J. B., Lee, M., Kesari, S., Abrouk, N. D., Day, R. M., Oronsky, A., Ray, C. M., Carterg, C. A. 2017; 26 (1): 109-119

    Abstract

    According to Hanahan and Weinberg, cancer manifests as six essential physiologic hallmarks: (1) self-sufficiency in growth signals, (2) insensitivity to growth-inhibitory signals, (3) evasion of programmed cell death, (4) limitless replicative potential, (5) sustained angiogenesis, and (6) invasion and metastasis. As a facilitator of these traits as well as immunosuppression and chemoresistance, the presence of tumor-associated macrophages (TAMs) may serve as the seventh hallmark of cancer. Anticancer agents that successfully reprogram TAMs to target rather than support tumor cells may hold the key to better therapeutic outcomes. Areas covered: This article summarizes the characteristics of the macrophage-stimulating agent RRx-001, a molecular iconoclast, sourced from the aerospace industry, with a particular emphasis on the cell-to-cell transfer mechanism of action (RBCs to TAMs) underlying its antitumor activity as well as its chemo and radioprotective properties, consolidated from various preclinical and clinical studies. Expert opinion: RRx-001 is macrophage-stimulating agent with the potential to synergize with chemotherapy, radiotherapy and immunotherapy while simultaneously protecting normal tissues from their cytotoxic effects. Given the promising indications of activity in multiple tumor types and these normal tissue protective properties, RRx-001 may be used to treat a broad spectrum of malignancies, if it is approved in the future.

    View details for DOI 10.1080/13543784.2017.1268600

    View details for PubMedID 27935336

  • Cabozantinib inhibits tumor growth and metastasis of a patient-derived xenograft model of papillary renal cell carcinoma with MET mutation CANCER BIOLOGY & THERAPY Zhao, H., Nolley, R., Chan, A. W., Rankin, E. B., Peehl, D. M. 2017; 18 (11): 863–71
  • Cabozantinib inhibits tumor growth and metastasis of a patient-derived xenograft model of papillary renal cell carcinoma with MET mutation. Cancer biology & therapy Zhao, H., Nolley, R., Chan, A. M., Rankin, E. B., Peehl, D. M. 2016: 0-?

    Abstract

    MET plays an important role in the development and progression of papillary renal cell carcinoma (pRCC). Evaluation of efficacy of MET inhibitors against pRCC has been hampered by limited preclinical models depicting MET abnormalities. We established a new patient-derived xenograft (PDX) model of pRCC carrying an activating mutation of MET and tested the ability of cabozantinib, an inhibitor of receptor tyrosine kinases including MET, to inhibit tumor growth and metastasis. Precision-cut, thin tissue slices from a pRCC specimen obtained by nephrectomy were implanted under the renal capsule of RAG2(-/-)γC(-/-) mice to establish first generation TSG-RCC-030. Histologic and genetic fidelity and metastatic potential of this model were characterized by immunohistochemistry, direct DNA sequencing and quantitative polymerase chain reaction (qPCR). The effect of cabozantinib on tumor growth and metastasis was evaluated. Whether measurements of circulating tumor DNA (ctDNA) by allele-specific qPCR could be used as a biomarker of tumor growth and response to therapy was determined. Subrenal and subcutaneous tumor grafts showed high take rates and metastasized to the lung. Both primary tumors and metastases expressed typical markers of pRCC and carried the same activating MET mutation as the parental tumor. Cabozantinib treatment caused striking tumor regression and inhibited lung metastasis in TSG-RCC-030. Plasma ctDNA levels correlated with tumor volume in control mice and changed in response to cabozantinib treatment. TSG-RCC-030 provides a realistic preclinical model to better understand the development and progression of pRCC with MET mutation and accelerate the development of new therapies for pRCC.

    View details for PubMedID 27715452

  • RRx-001, A novel dinitroazetidine radiosensitizer INVESTIGATIONAL NEW DRUGS Oronsky, B., Scicinski, J., Ning, S., Peehl, D., Oronsky, A., Cabrales, P., Bednarski, M., Knox, S. 2016; 34 (3): 371-377

    Abstract

    The 'holy grail' in radiation oncology is to improve the outcome of radiation therapy (RT) with a radiosensitizer-a systemic chemical/biochemical agent that additively or synergistically sensitizes tumor cells to radiation in the absence of significant toxicity. Similar to the oxygen effect, in which DNA bases modified by reactive oxygen species prevent repair of the cellular radiation damage, these compounds in general magnify free radical formation, leading to the permanent "fixation" of the resultant chemical change in the DNA structure. The purpose of this review is to present the origin story of the radiosensitizer, RRx-001, which emerged from the aerospace industry. The activity of RRx-001 as a chemosensitizer in multiple tumor types and disease states including malaria, hemorrhagic shock and sickle cell anemia, are the subject of future reviews.

    View details for DOI 10.1007/s10637-016-0326-y

    View details for Web of Science ID 000376129000012

    View details for PubMedID 26841903

    View details for PubMedCentralID PMC4859863

  • Spheroid culture of LuCaP 136 patient-derived xenograft enables versatile preclinical models of prostate cancer CLINICAL & EXPERIMENTAL METASTASIS Valta, M. P., Zhao, H., Saar, M., Tuomela, J., Nolley, R., Linxweiler, J., Sandholm, J., Lehtimaki, J., Harkonen, P., Coleman, I., Nelson, P. S., Corey, E., Peehl, D. M. 2016; 33 (4): 325-337

    Abstract

    LuCaP serially transplantable patient-derived xenografts (PDXs) are valuable preclinical models of locally advanced or metastatic prostate cancer. Using spheroid culture methodology, we recently established cell lines from several LuCaP PDXs. Here, we characterized in depth the features of xenografts derived from LuCaP 136 spheroid cultures and found faithful retention of the phenotype of the original PDX. In vitro culture enabled luciferase transfection into LuCaP 136 spheroids, facilitating in vivo imaging. We showed that LuCaP 136 spheroids formed intratibial, orthotopic, and subcutaneous tumors when re-introduced into mice. Intratibial tumors responded to castration and were highly osteosclerotic. LuCaP 136 is a realistic in vitro-in vivo preclinical model of a subtype of bone metastatic prostate cancer.

    View details for DOI 10.1007/s10585-016-9781-2

    View details for Web of Science ID 000373005900003

  • A Chemical Glycoproteomics Platform To study the Human Prostate Secretome Spiciarich, D. R., Purcell, S., Maund, S. L., Peehl, D. M., Bertozzi, C. R. FEDERATION AMER SOC EXP BIOL. 2016
  • Rockets, Radiosensitizers, and RRx-001: An Origin Story Part I DISCOVERY MEDICINE Oronsky, B., Scicinski, J., Ning, S., Peehl, D., Oronsky, A., Cabrales, P., Bednarski, M., Knox, S. 2016; 115: 173-180

    Abstract

    From Adam and Eve, to Darwinism, origin stories attempt to fill in the blanks, connect the dots, and define the turning points that are fundamental to subsequent developments. The purpose of this review is to present the origin story of a one-of-a-kind anticancer agent, RRx-001, which emerged from the aerospace industry as a putative radiosensitizer; not since the dynamite-to-dilator transformation of nitroglycerin in 1878 or the post-World War II explosive-to-elixir conversion of hydralazine, an ingredient in rocket fuel, to an antihypertensive, an antidepressant and an antituberculant, has energetic chemistry been harnessed for therapeutic purposes. This is Part 1 of the radiosensitization story; Parts 2 and 3, which detail the crossover activity of RRx-001 as a chemosensitizer in multiple tumor types and disease states including malaria, hemorrhagic shock and sickle cell anemia, are the subject of future reviews.

    View details for Web of Science ID 000376406200003

  • Non-invasive differentiation of benign renal tumors from clear cell renal cell carcinomas using clinically translatable hyperpolarized (13)C pyruvate magnetic resonance. Tomography : a journal for imaging research Sriram, R., Van Criekinge, M., Delos Santos, J., Keshari, K. R., Wilson, D. M., Peehl, D., Kurhanewicz, J., Wang, Z. J. 2016; 2 (1): 35-42

    Abstract

    Localized renal tumors are increasingly detected incidentally at imaging. Conventional imaging cannot reliably differentiate the 20% of these tumors that are benign from malignant renal cell carcinomas (RCCs), leading to unnecessary surgical resection and resulting morbidity associated with surgery. Here, we investigated hyperpolarized (13)C pyruvate metabolism in live patient-derived renal tumor tissue slices using a novel magnetic resonance (MR) -compatible bioreactor platform. We demonstrated for the first time that clear cell RCCs (ccRCCs), which account for 70-80% of all RCCs, have increased lactate production as well as rapid lactate efflux compared to benign renal tumors. This difference is attributed to increased lactate dehydrogenase A and monocarboxylate transporter 4 expression in ccRCCs. This distinctive metabolic phenotype can be used to differentiate RCCs from benign renal tumors using clinically translatable hyperpolarized (13)C pyruvate MR.

    View details for PubMedID 27227168

  • Endoscopic detection of cancer with lensless radioluminescence imaging and machine vision. Scientific reports Türkcan, S., Naczynski, D. J., Nolley, R., Sasportas, L. S., Peehl, D. M., Pratx, G. 2016; 6: 30737-?

    Abstract

    Complete removal of residual tumor tissue during surgical resection improves patient outcomes. However, it is often difficult for surgeons to delineate the tumor beyond its visible boundary. This has led to the development of intraoperative detectors that can image radiotracers accumulated within tumors, thus facilitating the removal of residual tumor tissue during surgical procedures. We introduce a beta imaging system that converts the beta radiation from the radiotracer into photons close to the decay origin through a CdWO4 scintillator and does not use any optical elements. The signal is relayed onto an EMCCD chip through a wound imaging fiber. The sensitivity of the device allows imaging of activity down to 100 nCi and the system has a resolution of at least 500 μm with a field of view of 4.80 × 6.51 mm. Advances in handheld beta cameras have focused on hardware improvements, but we apply machine vision to the recorded images to extract more information. We automatically classify sample regions in human renal cancer tissue ex-vivo into tumor or benign tissue based on image features. Machine vision boosts the ability of our system to distinguish tumor from healthy tissue by a factor of 9 ± 3 and can be applied to other beta imaging probes.

    View details for DOI 10.1038/srep30737

    View details for PubMedID 27477912

    View details for PubMedCentralID PMC4967900

  • Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. eLife Zhao, H., Yang, L., Baddour, J., Achreja, A., Bernard, V., Moss, T., Marini, J. C., Tudawe, T., Seviour, E. G., San Lucas, F. A., Alvarez, H., Gupta, S., Maiti, S. N., Cooper, L., Peehl, D., Ram, P. T., Maitra, A., Nagrath, D. 2016; 5

    Abstract

    Cancer-associated fibroblasts (CAFs) are a major cellular component of tumor microenvironment in most solid cancers. Altered cellular metabolism is a hallmark of cancer, and much of the published literature has focused on neoplastic cell-autonomous processes for these adaptations. We demonstrate that exosomes secreted by patient-derived CAFs can strikingly reprogram the metabolic machinery following their uptake by cancer cells. We find that CAF-derived exosomes (CDEs) inhibit mitochondrial oxidative phosphorylation, thereby increasing glycolysis and glutamine-dependent reductive carboxylation in cancer cells. Through 13C-labeled isotope labeling experiments we elucidate that exosomes supply amino acids to nutrient-deprived cancer cells in a mechanism similar to macropinocytosis, albeit without the previously described dependence on oncogenic-Kras signaling. Using intra-exosomal metabolomics, we provide compelling evidence that CDEs contain intact metabolites, including amino acids, lipids, and TCA-cycle intermediates that are avidly utilized by cancer cells for central carbon metabolism and promoting tumor growth under nutrient deprivation or nutrient stressed conditions.

    View details for DOI 10.7554/eLife.10250

    View details for PubMedID 26920219

  • A Protective Role for Androgen Receptor in Clear Cell Renal Cell Carcinoma Based on Mining TCGA Data. PloS one Zhao, H., Leppert, J. T., Peehl, D. M. 2016; 11 (1)

    Abstract

    Androgen receptor (AR) is expressed in normal murine and human kidneys of both genders, but its physiologic role is uncertain. Several studies showed loss of AR in renal cell carcinoma (RCC) in conjunction with increasing clinical stage and pathological grade, but others found that higher AR expression correlated with worse outcomes. Limited functional studies with renal cell lines suggested tumor-promoting activity of AR. In this study, we queried transcriptomic, proteomic, epigenetic and survival data from The Cancer Genome Atlas (TCGA) to evaluate AR expression and its association with overall survival in three subtypes of RCC (clear cell [ccRCC], papillary [pRCC], and chromophobe [chRCC]). We found that although there was no significant difference in AR mRNA expression in ccRCC of males vs. females, AR protein expression in ccRCC was significantly higher in male compared to female patients. More importantly, higher expression of AR at both transcript and protein levels was associated with improved overall survival in both genders with ccRCC, but did not predict survival of either gender with pRCC or chRCC. Genes whose transcript levels were associated with AR mRNA levels significantly overlapped between ccRCC and pRCC, but not with chRCC, suggesting a similar transcriptional program mediated by AR in ccRCC and pRCC. Ingenuity pathway analysis also identified overlapping pathways and upstream regulators enriched in AR-associated genes in ccRCC and pRCC. Hypermethylation of CpG sites located in the promoter and first exon of AR was associated with loss of AR expression and poor overall survival. Our findings support a tumor suppressor role for AR in both genders that might be exploited to decrease the incidence or progression of ccRCC.

    View details for DOI 10.1371/journal.pone.0146505

    View details for PubMedID 26814892

    View details for PubMedCentralID PMC4729482

  • Epigenetic effects of RRx-001: a possible unifying mechanism of anticancer activity. Oncotarget Zhao, H., Ning, S., Scicinski, J., Oronsky, B., Knox, S. J., Peehl, D. M. 2015; 6 (41): 43172-43181

    Abstract

    RRx-001 is a novel aerospace-derived compound currently under investigation in several ongoing Phase II studies. In a Phase I trial, it demonstrated anti-cancer activity and evidence of resensitization to formerly effective therapies in heavily pre-treated patients with relapsed/refractory solid tumors. RRx-001 generates reactive oxygen and nitrogen species (ROS and RNS) and nitric oxide (NO), elicits changes in intracellular redox status, modulates tumor blood flow, hypoxia and vascular function and triggers apoptosis in cancer cells. We investigated the effect of RRx-001 on the epigenome of SCC VII cancer cells. RRx-001 at 0.5 and 2 μM significantly decreased global DNA methylation, i.e., 5-methylcytosine levels, in SCC VII cells. Consistently, 0.5-5 μM RRx-001 significantly decreased Dnmt1 and Dnmt3a protein expression in a dose- and time-dependent manner. In addition, global methylation profiling identified differentially methylated genes in SCC VII cells treated with 0.5, 2, and 5 μM RRx-001 compared to control cells. Twenty-three target sites were hypomethylated and 22 hypermethylated by >10% in the presence of at least two different concentrations of RRx-001. Moreover, RRx-001 at 2 μM significantly increased global acetylated histone H3 and H4 levels in SCC VII cells after 24 hour treatment, suggesting that RRx-001 regulates global acetylation in cancer cells. These results demonstrate that, in contrast to the traditional "one drug one target" paradigm, RRx-001 has multi(epi)target features, which contribute to its anti-cancer activity and may rationalize the resensitization to previously effective therapies observed in clinical trials and serve as a unifying mechanism for its anticancer activity.

    View details for DOI 10.18632/oncotarget.6526

    View details for PubMedID 26657731

    View details for PubMedCentralID PMC4791224

  • NO to cancer: The complex and multifaceted role of nitric oxide and the epigenetic nitric oxide donor, RRx-001 REDOX BIOLOGY Scicinski, J., Oronsky, B., Ning, S., Knox, S., Peehl, D., Kim, M. M., Langecker, P., Fanger, G. 2015; 6: 1-8

    Abstract

    The endogenous mediator of vasodilation, nitric oxide (NO), has been shown to be a potent radiosensitizer. However, the underlying mode of action for its role as a radiosensitizer - while not entirely understood - is believed to arise from increased tumor blood flow, effects on cellular respiration, on cell signaling, and on the production of reactive oxygen and nitrogen species (RONS), that can act as radiosensitizers in their own right. NO activity is surprisingly long-lived and more potent in comparison to oxygen. Reports of the effects of NO with radiation have often been contradictory leading to confusion about the true radiosensitizing nature of NO. Whether increasing or decreasing tumor blood flow, acting as radiosensitizer or radioprotector, the effects of NO have been controversial. Key to understanding the role of NO as a radiosensitizer is to recognize the importance of biological context. With a very short half-life and potent activity, the local effects of NO need to be carefully considered and understood when using NO as a radiosensitizer. The systemic effects of NO donors can cause extensive side effects, and also affect the local tumor microenvironment, both directly and indirectly. To minimize systemic effects and maximize effects on tumors, agents that deliver NO on demand selectively to tumors using hypoxia as a trigger may be of greater interest as radiosensitizers. Herein we discuss the multiple effects of NO and focus on the clinical molecule RRx-001, a hypoxia-activated NO donor currently being investigated as a radiosensitizer in the clinic.

    View details for DOI 10.1016/j.redox.2015.07.002

    View details for Web of Science ID 000367338700001

    View details for PubMedCentralID PMC4529402

  • NO to cancer: The complex and multifaceted role of nitric oxide and the epigenetic nitric oxide donor, RRx-001. Redox biology Scicinski, J., Oronsky, B., Ning, S., Knox, S., Peehl, D., Kim, M. M., Langecker, P., Fanger, G. 2015; 6: 1-8

    Abstract

    The endogenous mediator of vasodilation, nitric oxide (NO), has been shown to be a potent radiosensitizer. However, the underlying mode of action for its role as a radiosensitizer - while not entirely understood - is believed to arise from increased tumor blood flow, effects on cellular respiration, on cell signaling, and on the production of reactive oxygen and nitrogen species (RONS), that can act as radiosensitizers in their own right. NO activity is surprisingly long-lived and more potent in comparison to oxygen. Reports of the effects of NO with radiation have often been contradictory leading to confusion about the true radiosensitizing nature of NO. Whether increasing or decreasing tumor blood flow, acting as radiosensitizer or radioprotector, the effects of NO have been controversial. Key to understanding the role of NO as a radiosensitizer is to recognize the importance of biological context. With a very short half-life and potent activity, the local effects of NO need to be carefully considered and understood when using NO as a radiosensitizer. The systemic effects of NO donors can cause extensive side effects, and also affect the local tumor microenvironment, both directly and indirectly. To minimize systemic effects and maximize effects on tumors, agents that deliver NO on demand selectively to tumors using hypoxia as a trigger may be of greater interest as radiosensitizers. Herein we discuss the multiple effects of NO and focus on the clinical molecule RRx-001, a hypoxia-activated NO donor currently being investigated as a radiosensitizer in the clinic.

    View details for DOI 10.1016/j.redox.2015.07.002

    View details for PubMedID 26164533

    View details for PubMedCentralID PMC4529402

  • Metabolic response of prostate cancer to nicotinamide phophoribosyltransferase inhibition in a hyperpolarized MR/PET compatible bioreactor PROSTATE Keshari, K. R., Wilson, D. M., Van Criekinge, M., Sriram, R., Koelsch, B. L., Wang, Z. J., VanBrocklin, H. F., Peehl, D. M., O'Brien, T., Sampath, D., Carano, R. A., Kurhanewicz, J. 2015; 75 (14): 1601-1609

    Abstract

    Metabolic shifts in disease are of great interest for the development of novel therapeutics. In cancer treatment, these therapies exploit the metabolic phenotype associated with oncogenesis and cancer progression. One recent strategy involves the depletion of the cofactors needed to maintain the high rate of glycolysis seen with the Warburg effect. Specifically, blocking nicotinamide adenine dinucleotide (NAD) biosynthesis via nicotinamide phosphoribosyltransferase (NAMPT) inhibition depletes cancer cells of the NAD needed for glycolysis. To characterize this metabolic phenotype in vivo and describe changes in flux with treatment, non-invasive biomarkers are necessary. One such biomarker is hyperpolarized (HP) [1-(13) C] pyruvate, a clinically translatable probe that allows real-time assessment of metabolism.We therefore developed a cell perfusion system compatible with HP magnetic resonance (MR) and positron emission tomography (PET) to develop translatable biomarkers of response to NAMPT inhibition in reduced volume cell cultures.Using this platform, we observed a reduction in pyruvate flux through lactate dehydrogenase with NAMPT inhibition in prostate cancer cells, and showed that both HP lactate and 2-[(18) F] fluoro-2-deoxy-D-glucose (FDG) can be used as biomarkers for treatment response of such targeted agents. Moreover, we observed dynamic flux changes whereby HP pyruvate was re-routed to alanine, providing both positive and negative indicators of treatment response.This study demonstrated the feasibility of a MR/PET compatible bioreactor approach to efficiently explore cell and tissue metabolism, the understanding of which is critical for developing clinically translatable biomarkers of disease states and responses to therapeutics. Prostate 75:1601-1609, 2015. © 2015 Wiley Periodicals, Inc.

    View details for DOI 10.1002/pros.23036

    View details for PubMedID 26177608

  • Safety and activity of RRx-001 in patients with advanced cancer: a first-in-human, open-label, dose-escalation phase 1 study LANCET ONCOLOGY Reid, T., Oronsky, B., Scicinski, J., Scribner, C. L., Knox, S. J., Ning, S., Peehl, D. M., Korn, R., Stirn, M., Carter, C. A., Oronsky, A., Taylor, M. J., Fitch, W. L., Cabrales, P., Kim, M. M., Burris, H. A., Lao, C. D., Abrouk, N. E., Fanger, G. R., Infante, J. R. 2015; 16 (9): 1133-1142

    Abstract

    Epigenetic alterations have been strongly associated with tumour formation and resistance to chemotherapeutic drugs, and epigenetic modifications are an attractive target in cancer research. RRx-001 is activated by hypoxia and induces the generation of reactive oxygen and nitrogen species that can epigenetically modulate DNA methylation, histone deacetylation, and lysine demethylation. The aim of this phase 1 study was to assess the safety, tolerability, and pharmacokinetics of RRx-001.In this open-label, dose-escalation, phase 1 study, we recruited adult patients (aged >18 years) with histologically or cytologically confirmed diagnosis of advanced, malignant, incurable solid tumours from University of California at San Diego, CA, USA, and Sarah Cannon Research Institute, Nashville, TN, USA. Key eligibility criteria included evaluable disease, Eastern Cooperative Group performance status of 2 or less, an estimated life expectancy of at least 12 weeks, adequate laboratory parameters, discontinuation of all previous antineoplastic therapies at least 6 weeks before intervention, and no residual side-effects from previous therapies. Patients were assigned to receive intravenous infusions of RRx-001 at increasing doses (10 mg/m(2), 16·7 mg/m(2), 24·6 mg/m(2), 33 mg/m(2), 55 mg/m(2), and 83 mg/m(2)) either once or twice-weekly for at least 4 weeks, with at least three patients per dose cohort and allowing a 2-week observation period before dose escalation. Samples for safety and pharmacokinetics analysis, including standard chemistry and haematological panels, were taken on each treatment day. The primary objective was to assess safety, tolerability, and dose-limiting toxic effects of RRx-001, to determine single-dose pharmacokinetics, and to identify a recommended dose for phase 2 trials. All analyses were done per protocol. Accrual is complete and follow-up is still on-going. This trial is registered with ClinicalTrials.gov, number NCT01359982.Between Oct 10, 2011, and March 18, 2013, we enrolled 25 patients and treated six patients in the 10 mg/m(2) cohort, three patients in the 16·7 mg/m(2) cohort, three patients in the 24·6 mg/m(2) cohort, four patients in the 33 mg/m(2) cohort, three patients in the 55 mg/m(2), and six patients in the 83 mg/m(2) cohort. Pain at the injection site, mostly grade 1 and grade 2, was the most common adverse event related to treatment, experienced by 21 (84%) patients. Other common drug-related adverse events included arm swelling or oedema (eight [32%] patients), and vein hardening (seven [28%] patients). No dose-limiting toxicities were observed. Time constraints related to management of infusion pain from RRx-001 resulted in a maximally feasible dose of 83 mg/m(2). Of the 21 evaluable patients, one (5%) patient had a partial response, 14 (67%) patients had stable disease, and six (29%) patients had progressive disease; all responses were across a variety of tumour types. Four patients who had received RRx-001 were subsequently rechallenged with a treatment that they had become refractory to; all four responded to the rechallenge.RRx-001 is a well-tolerated novel compound without clinically significant toxic effects at the tested doses. Preliminary evidence of activity is promising and, on the basis of all findings, a dose of 16·7 mg/m(2) was recommended as the targeted dose for phase 2 trials.EpicentRx (formerly RadioRx).

    View details for DOI 10.1016/S1470-2045(15)00089-3

    View details for PubMedID 26296952

  • Nrf2 activity as a potential biomarker for the pan-epigenetic anticancer agent, RRx-001. Oncotarget Ning, S., Sekar, T. V., Scicinski, J., Oronsky, B., Peehl, D. M., Knox, S. J., Paulmurugan, R. 2015; 6 (25): 21547-21556

    Abstract

    Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulatory transcription factor that plays an important role in the antioxidant response pathway against anticancer drug-induced cytotoxic effects. RRx-001 is a new anticancer agent that generates reactive oxygen and nitrogen species, and leads to epigenetic alterations in cancer cells. Here we report the RRx-001 mediated nuclear translocation of Nrf2 and the activation of expression of its downstream enzymes HO-1 and NQO1 in tumor cells. Inhibition of intrinsic Nrf2 expression by Nrf2-specific siRNA increased cell sensitivity to RRx-001. Molecular imaging of tumor cells co-expressing pARE-Firefly luciferase and pCMV-Renilla luciferase-mRFP in vitro and in vivo in mice revealed that RRx-001 significantly increased ARE-FLUC signal in cells in a dose- and time-dependent manner, suggesting that RRx-001 is an effective activator of the Nrf2-ARE signaling pathway. The pre-treatment level of ARE-FLUC signal in cells, reflecting basal activity of Nrf2, negatively correlated with the tumor response to RRx-001. The results support the concept that RRx-001 activates Nrf2-ARE antioxidant signaling pathways in tumor cells. Hence measurement of Nrf2-mediated activation of downstream target genes through ARE signaling may constitute a useful molecular biomarker for the early prediction of response to RRx-001 treatment, and thereby guide therapeutic decision-making.

    View details for PubMedID 26280276

  • Probing the prostate cancer secretome: Biomarker identification via bioorthogonal chemistry and MS proteomics Purcell, S., Spiciarich, D., Maund, S., Peehl, D., Bertozzi, C. AMER CHEMICAL SOC. 2015
  • RRx-001: A double action systemically non-toxic epigenetic agent for cancer therapy Zhao, H., Ning, S., Scicinski, J., Oronsky, B., Knox, S., Peehl, D. M. AMER ASSOC CANCER RESEARCH. 2015
  • Patient-derived prostate cancer cells for piloting of drug sensitivity and resistance testing Saeed, K., Rahkama, V., Eldfors, S., Bychkov, D., Mpindi, J., Yadav, B., Paavolainen, L., Aittokallio, T., Wennerberg, K., Peehl, D. M., Horvath, P., Mirtti, T., Rannikko, A., Kallioniemi, O., Ostling, P., af Hallstrom, T. M. WILEY-BLACKWELL. 2015: 49
  • Hepcidin Regulation in Prostate and Its Disruption in Prostate Cancer CANCER RESEARCH Tesfay, L., Clausen, K. A., Kim, J. W., Hegde, P., Wang, X., Miller, L. D., Deng, Z., Blanchette, N., Arvedson, T., Miranti, C. K., Babitt, J. L., Lin, H. Y., Peehl, D. M., Torti, F. M., Torti, S. V. 2015; 75 (11): 2254-2263

    Abstract

    Hepcidin is a circulating peptide hormone made by the liver that is a central regulator of systemic iron uptake and recycling. Here, we report that prostate epithelial cells also synthesize hepcidin, and that synthesis and secretion of hepcidin are markedly increased in prostate cancer cells and tissue. Prostatic hepcidin functions as an autocrine hormone, decreasing cell surface ferroportin, an iron exporter, increasing intracellular iron retention, and promoting prostate cancer cell survival. Synthesis of hepcidin in prostate cancer is controlled by a unique intersection of pathways that involves BMP4/7, IL6, Wnt, and the dual BMP and Wnt antagonist, SOSTDC1. Epigenetic silencing of SOSTDC1 through methylation is increased in prostate cancer and is associated with accelerated disease progression in patients with prostate cancer. These results establish a new connection between iron metabolism and prostate cancer, and suggest that prostatic dysregulation of hepcidin contributes to prostate cancer growth and progression.

    View details for DOI 10.1158/0008-5472.CAN-14-2465

    View details for Web of Science ID 000357333500014

    View details for PubMedID 25858146

    View details for PubMedCentralID PMC4454355

  • Elevated Serum MicroRNA Levels Associate with Absence of High-Grade Prostate Cancer in a Retrospective Cohort PLOS ONE Mihelich, B. L., Maranville, J. C., Nolley, R., Peehl, D. M., Nonn, L. 2015; 10 (4)

    Abstract

    To reduce treatment of indolent prostate cancer (PCa), biomarkers are needed to improve identification of patients with a low-risk of having aggressive disease. Over-treatment of these patients occurs because of uncertainty in the aggressiveness of the entire tumor based on the biopsies, which do not accurately sample multifocal tumors. Circulating microRNAs (miRNAs) are stable serum markers and differential miRNA levels occur in men with PCa. The goal of this study was to identify circulating miRNAs that were associated with aggressive or indolent PCa. We measured circulating miRNAs in 150 patients prior to surgery and compared the miRNA levels to the pathology of the entire radical prostatectomy specimen. For this study we used an exceptionally well-characterized cohort of patients who had benign prostatic hyperplasia (BPH), low-grade or high-grade PCa. Low-grade was defined as patients with 100% Gleason grade 3 tumor as determined by step-wise sectioning. High-grade PCa patients had 30-90% Gleason grade 4+5 in the tumor. BPH patients had at least two biopsies negative for PCa. Twenty one miRNAs were selected for analysis. The miRNAs were quantified by RT-qPCR and analyzed by logistic regression. High levels of 14 miRNAs were exclusively present in the serum from patients with low-grade PCa or BPH, compared to men with high-grade PCa who had consistently low levels. The expression levels of the 14 miRNAs were combined into a "miR Score" which had a negative predictive value (NPV) of 0.939 to predict absence of high-grade PCa among PCa and BPH patients. Biochemical recurrence (BCR) was known for the PCa patients and a combined "miR Risk Score" accurately classified a subset of patients with low risk of BCR (NPV 0.941). In summary, measurement of serum miRNAs may have pre-surgical utility in combination with clinical risk calculators to identify patients with low risk of harboring aggressive PCa.

    View details for DOI 10.1371/journal.pone.0124245

    View details for Web of Science ID 000353014700061

    View details for PubMedID 25874774

    View details for PubMedCentralID PMC4396984

  • Coordinate Loss of MAP3K7 and CHD1 Promotes Aggressive Prostate Cancer CANCER RESEARCH Rodrigues, L. U., Rider, L., Nieto, C., Romero, L., Karimpour-Fard, A., Loda, M., Lucia, M. S., Wu, M., Shi, L., Cimic, A., Sirintrapun, S. J., Nolley, R., Pac, C., Chen, H., Peehl, D. M., Xu, J., Liu, W., Costello, J. C., Cramer, S. D. 2015; 75 (6): 1021-1034

    Abstract

    Prostate cancer subtypes are poorly defined and functional validation of drivers of ETS rearrangement-negative prostate cancer has not been conducted. Here, we identified an ETS(-) subtype of aggressive prostate cancer (ERG(-)MAP3K7(del)CHD1(del)) and used a novel developmental model and a cell line xenograft model to show that cosuppression of MAP3K7 and CHD1 expression promotes aggressive disease. Analyses of publicly available prostate cancer datasets revealed that MAP3K7 and CHD1 were significantly codeleted in 10% to 20% of localized tumors and combined loss correlated with poor disease-free survival. To evaluate the functional impact of dual MAP3K7-CHD1 loss, we suppressed Map3k7 and/or Chd1 expression in mouse prostate epithelial progenitor/stem cells (PrP/SC) and performed tissue recombination experiments in vivo. Dual shMap3k7-shChd1 PrP/SC recombinants displayed massive glandular atypia with regions of prostatic intraepithelial neoplasia and carcinoma apparent. Combined Map3k7-Chd1 suppression greatly disrupted normal prostatic lineage differentiation; dual recombinants displayed significant androgen receptor loss, increased neuroendocrine differentiation, and increased neural differentiation. Clinical samples with dual MAP3K7-CHD1 loss also displayed neuroendocrine and neural characteristics. In addition, dual Map3k7-Chd1 suppression promoted E-cadherin loss and mucin production in recombinants. MAP3K7 and CHD1 protein loss also correlated with Gleason grade and E-cadherin loss in clinical samples. To further validate the phenotype observed in the PrP/SC model, we suppressed MAP3K7 and/or CHD1 expression in LNCaP prostate cancer cells. Dual shMAP3K7-shCHD1 LNCaP xenografts displayed increased tumor growth and decreased survival compared with shControl, shMAP3K7, and shCHD1 xenografts. Collectively, these data identify coordinate loss of MAP3K7 and CHD1 as a unique driver of aggressive prostate cancer development.

    View details for DOI 10.1158/0008-5472.CAN-14-1596

    View details for Web of Science ID 000351941400012

    View details for PubMedID 25770290

  • Expression of the leukocyte chemoattractant chemerin in human prostate tumors. Pachynski, R., Zabel, B., Leong, W., Crowder, R., Peehl, D., Butcher, E. AMER SOC CLINICAL ONCOLOGY. 2015
  • A strategy for tissue self-organization that is robust to cellular heterogeneity and plasticity. Proceedings of the National Academy of Sciences of the United States of America Cerchiari, A. E., Garbe, J. C., Jee, N. Y., Todhunter, M. E., Broaders, K. E., Peehl, D. M., Desai, T. A., LaBarge, M. A., Thomson, M., Gartner, Z. J. 2015; 112 (7): 2287-2292

    Abstract

    Developing tissues contain motile populations of cells that can self-organize into spatially ordered tissues based on differences in their interfacial surface energies. However, it is unclear how self-organization by this mechanism remains robust when interfacial energies become heterogeneous in either time or space. The ducts and acini of the human mammary gland are prototypical heterogeneous and dynamic tissues comprising two concentrically arranged cell types. To investigate the consequences of cellular heterogeneity and plasticity on cell positioning in the mammary gland, we reconstituted its self-organization from aggregates of primary cells in vitro. We find that self-organization is dominated by the interfacial energy of the tissue-ECM boundary, rather than by differential homo- and heterotypic energies of cell-cell interaction. Surprisingly, interactions with the tissue-ECM boundary are binary, in that only one cell type interacts appreciably with the boundary. Using mathematical modeling and cell-type-specific knockdown of key regulators of cell-cell cohesion, we show that this strategy of self-organization is robust to severe perturbations affecting cell-cell contact formation. We also find that this mechanism of self-organization is conserved in the human prostate. Therefore, a binary interfacial interaction with the tissue boundary provides a flexible and generalizable strategy for forming and maintaining the structure of two-component tissues that exhibit abundant heterogeneity and plasticity. Our model also predicts that mutations affecting binary cell-ECM interactions are catastrophic and could contribute to loss of tissue architecture in diseases such as breast cancer.

    View details for DOI 10.1073/pnas.1410776112

    View details for PubMedID 25633040

    View details for PubMedCentralID PMC4343104

  • Patient-derived tissue culture model systems of renal cell carcinoma for development of clinically translatable metabolic biomarkers Sriram, R., Keshari, K. R., Van Criekinge, M., Kurhanewicz, J., Wilson, D. M., Peehl, D. M., Bok, R., Zhen, W. J. AMER ASSOC CANCER RESEARCH. 2014
  • Tumor microenvironment derived exosomes regulate prostate cancer metabolism Zhao, H., Yang, L., Achreja, A., Marini, J., Peehl, D., Nagrath, D. AMER ASSOC CANCER RESEARCH. 2014
  • Molecular imaging of RRx-001-induced oxidative stress in Nrf2-luciferase expressing SCC VII tumors in mice Ning, S., Sekar, T., Paulmurugan, R., Scicinski, J., Oronsky, B., Peehl, D., Knox, S. J. AMER ASSOC CANCER RESEARCH. 2014
  • Spheroid culture of LuCaP 147 as an authentic preclinical model of prostate cancer subtype with SPOP mutation and hypermutator phenotype CANCER LETTERS Saar, M., Zhao, H., Nolley, R., Young, S. R., Coleman, I., Nelson, P. S., Vessella, R. L., Peehl, D. M. 2014; 351 (2): 272-280

    Abstract

    LuCaP serially transplantable xenografts are valuable preclinical models of locally advanced or metastatic prostate cancer. For the first time, we recently succeeded in establishing and serially passaging spheroid cultures of several LuCaP xenografts. Here, we characterized in depth the molecular and cellular phenotype of LuCaP 147 cultures and found faithful retention of the characteristics of the original xenograft, including immunophenotype, genetic fidelity, gene expression profile and responsiveness to androgen. Furthermore, we demonstrated capabilities for high-throughput drug screening and that anti-cancer agents induced cell cycle arrest and apoptosis in spheroid cultures. Finally, we showed that cells formed tumors when re-introduced into mice, providing an authentic in vitro-in vivo preclinical model of a subtype of prostate cancer with a hypermutator phenotype and an SPOP mutation.

    View details for DOI 10.1016/j.canlet.2014.06.014

    View details for Web of Science ID 000340221800012

    View details for PubMedID 24998678

    View details for PubMedCentralID PMC4112013

  • Identifying prostate cancer biomarkers by profiling glycoproteins in human prostate tissue Spiciarich, D. R., Maund, S. L., Peehl, D. M., Bertozzi, C. R. AMER CHEMICAL SOC. 2014
  • Nuclear KLLN expression associates with improved relapse-free survival for prostate carcinoma. Endocrine-related cancer Wang, Y., Roma, A., Nolley, R., Abdul-Karim, F., Peehl, D. M., Eng, C. 2014; 21 (4): 579-586

    Abstract

    Men with organ-confined prostate cancer (CaP) are often treated with radical prostatectomy. Despite similar postoperative characteristics, a significant proportion of men with an intermediate risk of progression experience prostate-specific antigen (PSA)-defined failure, while others have relapse-free survival (RFS). Additional prognostic markers are needed to predict the outcome of these patients. KLLN is a transcription factor that regulates the cell cycle and induces apoptosis in cancer cells. We have shown that KLLN is an androgen-regulated gene and that loss of KLLN expression in primary CaP is associated with high Gleason score. In this retrospective study, we evaluated KLLN expression in the high-grade malignancy components from 109 men with intermediate risk CaP. Patients with nuclear KLLN-negative tumors had significantly higher preoperative serum PSA levels (12.24±2.37 ng/ml) and larger tumor volumes (4.61±0.71 cm(3)) compared with nuclear KLLN-positive patients (8.35±2.45 ng/ml, P=0.03, and 2.66±0.51 cm(3), P<0.0001, respectively). None of the nuclear KLLN-positive tumors had capsular penetration, whereas 34% of nuclear KLLN-negative tumors (P=0.004) had capsular penetration. Maintaining KLLN expression in tumor nuclei, but not in cytoplasm or stroma, associated with improved RFS after surgery (P=0.002). Only 7% of patients with nuclear KLLN-positive tumors had tumor recurrence, while 60% of patients in the KLLN-negative group developed PSA-defined failure with median relapse time of 6.6 months (P=0.0003). Our data suggest that KLLN expression may be used as a prognostic marker to predict outcome for intermediate risk patients, which could provide useful information for postoperative management.

    View details for DOI 10.1530/ERC-14-0148

    View details for PubMedID 24972837

  • Phosphorylcholine-coated semiconducting polymer nanoparticles as rapid and efficient labeling agents for in vivo cell tracking. Advanced healthcare materials Pu, K., Shuhendler, A. J., Valta, M. P., Cui, L., Saar, M., Peehl, D. M., Rao, J. 2014; 3 (8): 1292-1298

    Abstract

    Despite the pressing need to noninvasively monitor transplanted cells in vivo with fluorescence imaging, desirable fluorescent agents with rapid labeling capability, durable brightness, and ideal biocompatibility remain lacking. Here, phosphorylcholine-coated near-infrared (NIR) fluorescent semiconducting polymer nanoparticles (SPNs) are reported as a new class of rapid, efficient, and cytocompatible labeling nanoagents for in vivo cell tracking. The phosphorylcholine coating results in efficient and rapid endocytosis and allows the SPN to enter cells within 0.5 h in complete culture medium apparently independent of the cell type, while its NIR fluorescence leads to a tissue penetration depth of 0.5 cm. In comparison to quantum dots and Cy5.5, the SPN is tolerant to physiologically ubiquitous reactive oxygen species (ROS), resulting in durable fluorescence both in vitro and in vivo. These desirable physical and physiological properties of the SPN permit cell tracking of human renal cell carcinoma (RCC) cells in living mice at a lower limit of detection of 10 000 cells with no obvious alteration of cell phenotype after 12 d. SPNs thus can provide unique opportunities for optimizing cellular therapy and deciphering pathological processes as a cell tracking label.

    View details for DOI 10.1002/adhm.201300534

    View details for PubMedID 24668903

    View details for PubMedCentralID PMC4134769

  • Carbohydrate Sequence of the Prostate Cancer-associated Antigen F77 Assigned by a Mucin O-Glycome Designer Array JOURNAL OF BIOLOGICAL CHEMISTRY Gao, C., Liu, Y., Zhang, H., Zhang, Y., Fukuda, M. N., Palma, A. S., Kozak, R. P., Childs, R. A., Nonaka, M., Li, Z., Siegel, D. L., Hanfland, P., Peehl, D. M., Chai, W., Greene, M. I., Ten Feizi, F. Z. 2014; 289 (23): 16462-16477

    Abstract

    Monoclonal antibody F77 was previously raised against human prostate cancer cells and has been shown to recognize a carbohydrate antigen, but the carbohydrate sequence of the antigen was elusive. Here, we make multifaceted approaches to characterize F77 antigen, including binding analyses with the glycolipid extract of the prostate cancer cell line PC3, microarrays with sequence-defined glycan probes, and designer arrays from the O-glycome of an antigen-positive mucin, in conjunction with mass spectrometry. Our results reveal F77 antigen to be expressed on blood group H on a 6-linked branch of a poly-N-acetyllactosamine backbone. We show that mAb F77 can also bind to blood group A and B analogs but with lower intensities. We propose that the close association of F77 antigen with prostate cancers is a consequence of increased blood group H expression together with up-regulated branching enzymes. This is in contrast to other epithelial cancers that have up-regulated branching enzymes but diminished expression of H antigen. With knowledge of the structure and prevalence of F77 antigen in prostate cancer, the way is open to explore rationally its application as a biomarker to detect F77-positive circulating prostate cancer-derived glycoproteins and tumor cells.

    View details for DOI 10.1074/jbc.M114.558932

    View details for Web of Science ID 000337965700048

    View details for PubMedCentralID PMC4047413

  • Determination of Carbohydrate Structure Recognized by Prostate-specific F77 Monoclonal Antibody through Expression Analysis of Glycosyltransferase Genes. journal of biological chemistry Nonaka, M., Fukuda, M. N., Gao, C., Li, Z., Zhang, H., Greene, M. I., Peehl, D. M., Feizi, T., Fukuda, M. 2014; 289 (23): 16478-16486

    Abstract

    This study reports the determination of the carbohydrate epitope of monoclonal antibody F77 previously raised against human prostate cancer PC-3 cells (Zhang, G., Zhang, H., Wang, Q., Lal, P., Carroll, A. M., de la Llera-Moya, M., Xu, X., and Greene, M. I. (2010) Proc. Natl. Acad. Sci. U. S. A. 107, 732-737). We performed a series of co-transfections using mammalian expression vectors encoding specific glycosyltransferases. We thereby identified branching enzymes and FUT1 (required for Fucα1→2Gal linkage) as being essential for F77 antigen formation. When immortalized normal prostate 267B1 cells were transfected with FUT1 alone, cells showed weak expression of F77 antigen. By contrast, cells co-transfected with FUT1 plus either GCNT1, GCNT2, or GCNT3 (an enzyme required to form GlcNAcβ1→6Gal/GalNAc) showed robust F77 antigen expression, suggesting that F77 specifically binds to Fucα1→2Galβ1→4GlcNAcβ1→6Gal/GalNAc. RT-PCR for FUT1, GCNT1, GCNT2, and GCNT3 showed that F77-positive cell lines indeed express transcripts encoding FUT1 plus one GCNT. F77-positive prostate cancer cells transfected with siRNAs targeting FUT1, GCNT2, and GCNT3 showed significantly reduced F77 antigen, confirming the requirement of these enzymes for epitope synthesis. We also found that hypoxia induces F77 epitope expression in immortalized prostate RWPE1 cells, which express F77 antigen moderately under normoxia but at an elevated level under hypoxia. Quantitative RT-PCR demonstrated up-regulation of FUT1, GCNT2, and GCNT3 transcripts in RWPE1 cells under hypoxia, suggesting that hypoxia up-regulates glycosyltransferase expression required for F77 antigen synthesis. These results define the F77 epitope and provide a potential mechanism for F77 antigen synthesis in malignant prostate cancer.

    View details for DOI 10.1074/jbc.M114.559047

    View details for PubMedID 24753248

    View details for PubMedCentralID PMC4047414

  • Determination of Carbohydrate Structure Recognized by Prostate-specific F77 Monoclonal Antibody through Expression Analysis of Glycosyltransferase Genes JOURNAL OF BIOLOGICAL CHEMISTRY Nonaka, M., Fukuda, M. N., Gao, C., Li, Z., Zhang, H., Greene, M. I., Peehl, D. M., Ten Feizi, F. Z., Fukuda, M. 2014; 289 (23): 16478-16486

    Abstract

    This study reports the determination of the carbohydrate epitope of monoclonal antibody F77 previously raised against human prostate cancer PC-3 cells (Zhang, G., Zhang, H., Wang, Q., Lal, P., Carroll, A. M., de la Llera-Moya, M., Xu, X., and Greene, M. I. (2010) Proc. Natl. Acad. Sci. U. S. A. 107, 732-737). We performed a series of co-transfections using mammalian expression vectors encoding specific glycosyltransferases. We thereby identified branching enzymes and FUT1 (required for Fucα1→2Gal linkage) as being essential for F77 antigen formation. When immortalized normal prostate 267B1 cells were transfected with FUT1 alone, cells showed weak expression of F77 antigen. By contrast, cells co-transfected with FUT1 plus either GCNT1, GCNT2, or GCNT3 (an enzyme required to form GlcNAcβ1→6Gal/GalNAc) showed robust F77 antigen expression, suggesting that F77 specifically binds to Fucα1→2Galβ1→4GlcNAcβ1→6Gal/GalNAc. RT-PCR for FUT1, GCNT1, GCNT2, and GCNT3 showed that F77-positive cell lines indeed express transcripts encoding FUT1 plus one GCNT. F77-positive prostate cancer cells transfected with siRNAs targeting FUT1, GCNT2, and GCNT3 showed significantly reduced F77 antigen, confirming the requirement of these enzymes for epitope synthesis. We also found that hypoxia induces F77 epitope expression in immortalized prostate RWPE1 cells, which express F77 antigen moderately under normoxia but at an elevated level under hypoxia. Quantitative RT-PCR demonstrated up-regulation of FUT1, GCNT2, and GCNT3 transcripts in RWPE1 cells under hypoxia, suggesting that hypoxia up-regulates glycosyltransferase expression required for F77 antigen synthesis. These results define the F77 epitope and provide a potential mechanism for F77 antigen synthesis in malignant prostate cancer.

    View details for DOI 10.1074/jbc.M114.559047

    View details for Web of Science ID 000337965700049

    View details for PubMedCentralID PMC4047414

  • Carbohydrate sequence of the prostate cancer-associated antigen F77 assigned by a mucin O-glycome designer array. journal of biological chemistry Gao, C., Liu, Y., Zhang, H., Zhang, Y., Fukuda, M. N., Palma, A. S., Kozak, R. P., Childs, R. A., Nonaka, M., Li, Z., Siegel, D. L., Hanfland, P., Peehl, D. M., Chai, W., Greene, M. I., Feizi, T. 2014; 289 (23): 16462-16477

    Abstract

    Monoclonal antibody F77 was previously raised against human prostate cancer cells and has been shown to recognize a carbohydrate antigen, but the carbohydrate sequence of the antigen was elusive. Here, we make multifaceted approaches to characterize F77 antigen, including binding analyses with the glycolipid extract of the prostate cancer cell line PC3, microarrays with sequence-defined glycan probes, and designer arrays from the O-glycome of an antigen-positive mucin, in conjunction with mass spectrometry. Our results reveal F77 antigen to be expressed on blood group H on a 6-linked branch of a poly-N-acetyllactosamine backbone. We show that mAb F77 can also bind to blood group A and B analogs but with lower intensities. We propose that the close association of F77 antigen with prostate cancers is a consequence of increased blood group H expression together with up-regulated branching enzymes. This is in contrast to other epithelial cancers that have up-regulated branching enzymes but diminished expression of H antigen. With knowledge of the structure and prevalence of F77 antigen in prostate cancer, the way is open to explore rationally its application as a biomarker to detect F77-positive circulating prostate cancer-derived glycoproteins and tumor cells.

    View details for DOI 10.1074/jbc.M114.558932

    View details for PubMedID 24753245

    View details for PubMedCentralID PMC4047413

  • Development of a realistic in vivo bone metastasis model of human renal cell carcinoma CLINICAL & EXPERIMENTAL METASTASIS Valta, M. P., Zhao, H., Ingels, A., Thong, A. E., Nolley, R., Saar, M., Peehl, D. M. 2014; 31 (5): 573-584

    Abstract

    About one-third of patients with advanced renal cell carcinoma (RCC) have bone metastases. The incidence of RCC is increasing and bone metastatic RCC merits greater focus. Realistic preclinical bone metastasis models of RCC are lacking, hampering the development of effective therapies. We developed a realistic in vivo bone metastasis model of human RCC by implanting precision-cut tissue slices under the renal capsule of immunodeficient mice. The presence of disseminated cells in bone marrow of tissue slice graft (TSG)-bearing mice was screened by human-specific polymerase chain reaction and confirmed by immunohistology using human-specific antibody. Disseminated tumor cells in bone marrow of TSG-bearing mice derived from three of seven RCC patients were detected as early as 1 month after tissue implantation at a high frequency with close resemblance to parent tumors (e.g., CAIX expression and high vascularity). The metastatic patterns of TSGs correlated with disease progression in patients. In addition, TSGs retained capacity to metastasize to bone at high frequency after serial passaging and cryopreservation. Moreover, bone metastases in mice responded to Temsirolimus treatment. Intratibial injections of single cells generated from TSGs showed 100 % engraftment and produced X-ray-visible tumors as early as 3 weeks after cancer cell inoculation. Micro-computed tomography (μCT) and histological analysis revealed osteolytic characteristics of these lesions. Our results demonstrated that orthotopic RCC TSGs have potential to develop bone metastases that respond to standard therapy. This first reported primary RCC bone metastasis model provides a realistic setting to test therapeutics to prevent or treat bone metastases in RCC.

    View details for DOI 10.1007/s10585-014-9651-8

    View details for Web of Science ID 000337082400008

    View details for PubMedID 24715498

    View details for PubMedCentralID PMC4351963

  • Preclinical trial of a new dual mTOR inhibitor, MLN0128, using renal cell carcinoma tumorgrafts INTERNATIONAL JOURNAL OF CANCER Ingels, A., Zhao, H., Thong, A. E., Saar, M., Valta, M. P., Nolley, R., Santos, J., Peehl, D. M. 2014; 134 (10): 2322-2329

    Abstract

    mTOR is a rational target in renal cell carcinoma (RCC) because of its role in disease progression. However, the effects of temsirolimus, the only first-generation mTOR inhibitor approved by the FDA for first-line treatment of metastatic RCC, on tumor reduction and progression-free survival are minimal. Second-generation mTOR inhibitors have not been evaluated on RCC. We compared the effects of temsirolimus and MLN0128, a potent second-generation mTOR inhibitor, on RCC growth and metastasis using a realistic patient-derived tissue slice graft (TSG) model. TSGs were derived from three fresh primary RCC specimens by subrenal implantation of precision-cut tissue slices into immunodeficient mice that were randomized and treated with MLN0128, temsirolimus, or placebo. MLN0128 consistently suppressed primary RCC growth, monitored by magnetic resonance imaging (MRI), in three TSG cohorts for up to 2 months. Temsirolimus, in contrast, only transiently inhibited the growth of TSGs in one of two cohorts before resistance developed. In addition, MLN0128 reduced liver metastases, determined by human-specific quantitative polymerase chain reaction, in two TSG cohorts, whereas temsirolimus failed to have any significant impact. Moreover, MLN0128 decreased levels of key components of the two mTOR subpathways including TORC1 targets 4EBP1, p-S6K1, HIF1α and MTA1 and the TORC2 target c-Myc, consistent with dual inhibition. Our results demonstrated that MLN0128 is superior to temsirolimus in inhibiting primary RCC growth as well as metastases, lending strong support for further clinical development of dual mTOR inhibitors for RCC treatment.

    View details for DOI 10.1002/ijc.28579

    View details for Web of Science ID 000332309700007

    View details for PubMedID 24243565

    View details for PubMedCentralID PMC4365782

  • Identifying prostate cancer biomarkers by profiling glycoproteins in human prostate tissue Spiciarich, D., Maund, S., Peehl, D., Bertozzi, C. FEDERATION AMER SOC EXP BIOL. 2014
  • A Tissue Graft Model of DNA Damage Response in the Normal and Malignant Human Prostate JOURNAL OF UROLOGY af Hallstrom, T. M., Zhao, H., Tian, J., Rantanen, V., Reese, S. W., Nolley, R., Laiho, M., Peehl, D. M. 2014; 191 (3): 842-849

    Abstract

    DNA damage responses are relevant to prostate cancer initiation, progression and treatment. Few models of the normal and malignant human prostate that maintain stromal-epithelial interactions in vivo exist in which to study DNA damage responses. We evaluated the feasibility of maintaining tissue slice grafts at subcutaneous vs subrenal capsular sites in RAG2(-/-)γC(-/-) mice to study the DNA damage responses of normal and malignant glands.We compared the take rate and histology of tissue slice grafts from fresh, precision cut surgical specimens that were maintained for 1 to 4 weeks in subcutaneous vs subrenal capsular sites. Induction of γH2AX, p53, ATM and apoptosis was evaluated as a measure of the DNA damage response after irradiation.The take rate of subcutaneous tissue slice grafts was higher than typically reported but lower than at the subrenal capsular site. Subcutaneous tissue slice grafts frequently showed basal cell hyperplasia, squamous metaplasia and cystic atrophy, and cancer did not survive. In contrast, normal and malignant histology was well maintained in subrenal capsular tissue slice grafts. Regardless of implantation site the induction of γH2AX and ATM occurred in tissue slice graft epithelium 1 hour after irradiation and decreased to basal level by 24 hours, indicating DNA damage recognition and repair. As observed previously in prostatic ex vivo models, p53 was not activated. Notably, tumor but not normal cells responded to irradiation by undergoing apoptosis.To our knowledge this is the first study of DNA damage responses in a patient derived prostate tissue graft model. The subrenal capsular site of RAG2(-/-)γC(-/-) mice optimally maintains normal and malignant histology and function, permitting novel studies of DNA damage responses in a physiological context.

    View details for DOI 10.1016/j.juro.2013.09.007

    View details for Web of Science ID 000331123400095

    View details for PubMedID 24035881

    View details for PubMedCentralID PMC4009951

  • Optimization and comprehensive characterization of a faithful tissue culture model of the benign and malignant human prostate. Laboratory investigation; a journal of technical methods and pathology Maund, S. L., Nolley, R., Peehl, D. M. 2014; 94 (2): 208-221

    Abstract

    Few preclinical models accurately depict normal human prostate tissue or primary prostate cancer (PCa). In vitro systems typically lack complex cellular interactions among structured prostatic epithelia and a stromal microenvironment, and genetic and molecular fidelity are concerns in both in vitro and in vivo models. 'Tissue slice cultures' (TSCs) provide realistic preclinical models of diverse tissues and organs, but have not been fully developed or widely utilized for prostate studies. Problems encountered include degeneration of differentiated secretory cells, basal cell hyperplasia, and poor survival of PCa. Here, we optimized, characterized, and applied a TSC model of primary human PCa and benign prostate tissue that overcomes many deficiencies of current in vitro models. Tissue cores from fresh prostatectomy specimens were precision-cut at 300 μm and incubated in a rotary culture apparatus. The ability of varied culture conditions to faithfully maintain benign and cancer cell and tissue structure and function over time was evaluated by immunohistological and biochemical assays. After optimization of the culture system, molecular and cellular responses to androgen ablation and to piperlongumine (PL), purported to specifically reduce androgen signaling in PCa, were investigated. Optimized culture conditions successfully maintained the structural and functional fidelity of both benign and PCa TSCs for 5 days. TSCs exhibited androgen dependence, appropriately undergoing ductal degeneration, reduced proliferation, and decreased prostate-specific antigen expression upon androgen ablation. Further, TSCs revealed cancer-specific reduction of androgen receptor and increased apoptosis upon treatment with PL, validating data from cell lines. We demonstrate a TSC model that authentically recapitulates the structural, cellular, and genetic characteristics of the benign and malignant human prostate, androgen dependence of the native tissue, and cancer-specific response to a potentially new therapeutic for PCa. The work described herein provides a basis for advancing the experimental utility of the TSC model.

    View details for DOI 10.1038/labinvest.2013.141

    View details for PubMedID 24296879

  • Tissue slice grafts of human renal cell carcinoma: an authentic preclinical model with high engraftment rate and metastatic potential. Urologic oncology Thong, A. E., Zhao, H., Ingels, A., Valta, M. P., Nolley, R., Santos, J., Young, S. R., Peehl, D. M. 2014; 32 (1): 43 e23-30

    Abstract

    Discovery of curative therapies for renal cell carcinoma (RCC) is hampered by lack of authentic preclinical models. Tumorgrafts, generated by direct implantation of patient-derived tissues into mice, have demonstrated superior ability to predict therapeutic response. We evaluated "tissue slice grafts" (TSGs) as an improved tumorgraft model of RCC.Cores of fresh RCC were precision-cut at 300 µm and implanted under the renal capsule of RAG2(-/-)γC(-/-) mice. Engraftment rate, histology, biomarker expression, genetic fidelity, and metastatic potential were evaluated. Magnetic resonance imaging (MRI) was tested as a noninvasive method to measure tumor volume, and response to a targeted therapy was determined.All 13 cases of RCC engrafted and displayed characteristic histology and biomarkers. TSG volume quantified noninvasively by MRI highly correlated with graft weights, providing a unique tool for monitoring orthotopic growth. Moreover, in 2 cases, cancer cells from TSGs metastasized to clinically relevant sites, including bone. Microarray analysis and DNA sequencing demonstrated a high degree of correlation of global gene expression and von Hippel-Lindau (VHL) status between TSGs and parental tumors. Treatment of TSGs with sunitinib significantly decreased graft weight and mean vessel density compared with controls.The TSG model of RCC faithfully recapitulates tumor pathology, gene expression, genetic mutation, and drug response. The high engraftment rate and metastatic potential of this authentic model, in conjunction with the ability to generate large first-generation animal cohorts and to quantitate tumor volume at the orthotopic site by MRI, proffer significant advantages compared with other preclinical platforms.

    View details for DOI 10.1016/j.urolonc.2013.05.008

    View details for PubMedID 23911681

  • Tissue slice grafts of human renal cell carcinoma: An authentic preclinical model with high engraftment rate and metastatic potential UROLOGIC ONCOLOGY-SEMINARS AND ORIGINAL INVESTIGATIONS Thong, A. E., Zhao, H., Ingels, A., Valta, M. P., Nolley, R., Santos, J., Young, S. R., Peehl, D. M. 2014; 32 (1)

    Abstract

    Discovery of curative therapies for renal cell carcinoma (RCC) is hampered by lack of authentic preclinical models. Tumorgrafts, generated by direct implantation of patient-derived tissues into mice, have demonstrated superior ability to predict therapeutic response. We evaluated "tissue slice grafts" (TSGs) as an improved tumorgraft model of RCC.Cores of fresh RCC were precision-cut at 300 µm and implanted under the renal capsule of RAG2(-/-)γC(-/-) mice. Engraftment rate, histology, biomarker expression, genetic fidelity, and metastatic potential were evaluated. Magnetic resonance imaging (MRI) was tested as a noninvasive method to measure tumor volume, and response to a targeted therapy was determined.All 13 cases of RCC engrafted and displayed characteristic histology and biomarkers. TSG volume quantified noninvasively by MRI highly correlated with graft weights, providing a unique tool for monitoring orthotopic growth. Moreover, in 2 cases, cancer cells from TSGs metastasized to clinically relevant sites, including bone. Microarray analysis and DNA sequencing demonstrated a high degree of correlation of global gene expression and von Hippel-Lindau (VHL) status between TSGs and parental tumors. Treatment of TSGs with sunitinib significantly decreased graft weight and mean vessel density compared with controls.The TSG model of RCC faithfully recapitulates tumor pathology, gene expression, genetic mutation, and drug response. The high engraftment rate and metastatic potential of this authentic model, in conjunction with the ability to generate large first-generation animal cohorts and to quantitate tumor volume at the orthotopic site by MRI, proffer significant advantages compared with other preclinical platforms.

    View details for DOI 10.1016/j.urolonc.2013.05.008

    View details for Web of Science ID 000347243300079

    View details for PubMedID 23911681

    View details for PubMedCentralID PMC4350678

  • Imaging the Glycosylation State of Cell Surface Glycoproteins by Two-Photon Fluorescence Lifetime Imaging Microscopy ANGEWANDTE CHEMIE-INTERNATIONAL EDITION Belardi, B., de la Zerda, A., Spiciarich, D. R., Maund, S. L., Peehl, D. M., Bertozzi, C. R. 2013; 52 (52): 14045-14049

    View details for DOI 10.1002/anie.201307512

    View details for Web of Science ID 000328531100027

    View details for PubMedID 24259491

    View details for PubMedCentralID PMC3920747

  • Radioprotection and Cell Cycle Arrest of Intestinal Epithelial Cells by Darinaparsin, a Tumor Radiosensitizer INTERNATIONAL JOURNAL OF RADIATION ONCOLOGY BIOLOGY PHYSICS Tian, J., Doi, H., Saar, M., Santos, J., Li, X., Peehl, D. M., Knox, S. J. 2013; 87 (5): 1179-1185

    Abstract

    It was recently reported that the organic arsenic compound darinaparsin (DPS) is a cytotoxin and radiosensitizer of tumor cells in vitro and in subcutaneous xenograft tumors. Surprisingly, it was also found that DPS protects normal intestinal crypt epithelial cells (CECs) from clonogenic death after ionizing radiation (IR). Here we tested the DPS radiosensitizing effect in a clinically relevant model of prostate cancer and explored the radioprotective effect and mechanism of DPS on CECs.The radiation modification effect of DPS was tested in a mouse model of orthotopic xenograft prostate cancer and of IR-induced acute gastrointestinal syndrome. The effect of DPS on CEC DNA damage and DNA damage responses was determined by immunohistochemistry.In the mouse model of IR-induced gastrointestinal syndrome, DPS treatment before IR accelerated recovery from body weight loss and increased animal survival. DPS decreased post-IR DNA damage and cell death, suggesting that the radioprotective effect was mediated by enhanced DNA damage repair. Shortly after DPS injection, significant cell cycle arrest was observed in CECs at both G1/S and G2/M checkpoints, which was accompanied by the activation of cell cycle inhibitors p21 and growth arrest and DNA-damage-inducible protein 45 alpha (GADD45A). Further investigation revealed that DPS activated ataxia telangiectasia mutated (ATM), an important inducer of DNA damage repair and cell cycle arrest.DPS selectively radioprotected normal intestinal CECs and sensitized prostate cancer cells in a clinically relevant model. This effect may be, at least in part, mediated by DNA damage response activation and has the potential to significantly increase the therapeutic index of radiation therapy.

    View details for DOI 10.1016/j.ijrobp.2013.08.051

    View details for Web of Science ID 000327496600056

    View details for PubMedID 24210080

  • Establishment and serial passage of cell cultures derived from LuCaP xenografts PROSTATE Young, S. R., Saar, M., Santos, J., Nguyen, H. M., Vessella, R. L., Peehl, D. M. 2013; 73 (12): 1251-1262

    Abstract

    BACKGROUND: LuCaP serially transplantable xenografts derived from primary and metastatic human prostate cancer encompass the molecular and cellular heterogeneity of the disease and are an invaluable resource for in vivo preclinical studies. A limitation of this model, however, has been the inability to establish and passage cell cultures derived from the xenografts. Here, we describe a novel spheroid culture system that supports long-term growth of LuCaP cells in vitro. METHODS: Xenografts were minced and digested with collagenase. Tissue dissociation was terminated while the majority of cells remained as clusters rather than single cells. The cell clusters were suspended in StemPro medium supplemented with R1881 and Y-27632, a Rho kinase inhibitor, and placed in ultralow attachment dishes for spheroid culture. Serial passage was achieved by partial digestion to small clusters with trypsin/EDTA in the presence of Y-27632. Cell viability, growth and phenotype were monitored with LIVE/DEAD®, MTS, qRT-PCR, and immunocytochemical assays. RESULTS: Cells from six LuCaP xenografts formed proliferating spheroids that were serially passaged a minimum of three times and cryopreserved. Two of the cell lines, LuCaP 136 and LuCaP 147, were further passaged and characterized. Both expressed biomarkers characteristic of the xenografts of origin, were determined to be of independent origin by STR fingerprinting, and were free of mycoplasma. LuCaP 147 formed tumors similar to the original xenograft when injected into mice. CONCLUSIONS: The ability to culture LuCaP cells affords new opportunities for fast, cheap, and efficient preclinical studies and extends the value of the LuCaP xenograft models. Prostate 9999: XX-XX, 2013. © 2013 Wiley Periodicals, Inc.

    View details for DOI 10.1002/pros.22610

    View details for Web of Science ID 000322013100001

    View details for PubMedID 23740600

  • Patient-derived tissue slice grafts accurately depict response of high-risk primary prostate cancer to androgen deprivation therapy JOURNAL OF TRANSLATIONAL MEDICINE Zhao, H., Thong, A., Nolley, R., Reese, S. W., Santos, J., Ingels, A., Peehl, D. M. 2013; 11

    Abstract

    Effective eradication of high-risk primary prostate cancer (HRPCa) could significantly decrease mortality from prostate cancer. However, the discovery of curative therapies for HRPCa is hampered by the lack of authentic preclinical models.We improved upon tumorgraft models that have been shown to predict drug response in other cancer types by implanting thin, precision-cut slices of HRPCa under the renal capsule of immunodeficient mice. Tissue slice grafts (TSGs) from 6 cases of HRPCa were established in mice. Following androgen deprivation by castration, TSGs were recovered and the presence and phenotype of cancer cells were evaluated.High-grade cancer in TSGs generated from HRPCa displayed characteristic Gleason patterns and biomarker expression. Response to androgen deprivation therapy (ADT) was as in humans, with some cases exhibiting complete pathologic regression and others showing resistance to castration. As in humans, ADT decreased cell proliferation and prostate-specific antigen expression in TSGs. Adverse pathological features of parent HRPCa were associated with lack of regression of cancer in corresponding TSGs after ADT. Castration-resistant cancer cells remaining in TSGs showed upregulated expression of androgen receptor target genes, as occurs in castration-resistant prostate cancer (CRPC) in humans. Finally, a rare subset of castration-resistant cancer cells in TSGs underwent epithelial-mesenchymal transition, a process also observed in CRPC in humans.Our study demonstrates the feasibility of generating TSGs from multiple patients and of generating a relatively large number of TSGs from the same HRPCa specimen with similar cell composition and histology among control and experimental samples in an in vivo setting. The authentic response of TSGs to ADT, which has been extensively characterized in humans, suggests that TSGs can serve as a surrogate model for clinical trials to achieve rapid and less expensive screening of therapeutics for HRPCa and primary CRPC.

    View details for DOI 10.1186/1479-5876-11-199

    View details for Web of Science ID 000323850900001

    View details for PubMedID 23985008

    View details for PubMedCentralID PMC3766103

  • Metabolic Reprogramming and Validation of Hyperpolarized C-13 Lactate as a Prostate Cancer Biomarker Using a Human Prostate Tissue Slice Culture Bioreactor PROSTATE Keshari, K. R., Sriram, R., Van Criekinge, M., Wilson, D. M., Wang, Z. J., Vigneron, D. B., Peehl, D. M., Kurhanewicz, J. 2013; 73 (11): 1171-1181

    Abstract

    BACKGROUND: The treatment of prostate cancer has been impeded by the lack of both clinically relevant disease models and metabolic markers that track tumor progression. Hyperpolarized (HP) (13) C MR spectroscopy has emerged as a new technology to investigate the metabolic shifts in prostate cancer. In this study, we investigate the glucose reprogramming using HP (13) C pyruvate MR in a patient-derived prostate tissue slice culture (TSC) model. METHODS: The steady-state metabolite concentrations in freshly excised human prostate TSCs were assessed and compared to those from snap-frozen biopsy samples. The TSCs were then applied to a perfused cell (bioreactor) platform, and the bioenergetics and the dynamic pyruvate flux of the TSCs were investigated by (31) P and HP (13) C MR, respectively. RESULTS: The prostate TSCs demonstrated steady-state glycolytic and phospholipid metabolism, and bioenergetics that recapitulate features of prostate cancer in vivo. (13) C spectra following injection of HP (13) C pyruvate showed significantly increased pyruvate to lactate flux in malignant as compared to the benign prostate TSCs. This increased flux in the malignant prostate TSCs correlated with both increased expression of monocarboxylate transporters (MCT) and activity of lactate dehydrogenase (LDH). CONCLUSIONS: We provide the first mechanistic evidence for HP (13) C lactate as a prostate cancer biomarker in living human tissues, critical for the interpretation of in vivo studies. More broadly, the clinically relevant metabolic model system in combination with HP MR can facilitate the identification of clinically translatable biomarkers of prostate cancer presence, aggressiveness, and treatment response. Prostate © 2013 Wiley Periodicals, Inc.

    View details for DOI 10.1002/pros.22665

    View details for Web of Science ID 000330191800004

    View details for PubMedID 23532911

  • Patient-derived tissue slice grafts of high-risk primary prostate cancer: An authentic preclinical model for synthetic lethality-based therapy Zhao, H., Thong, A., Nolley, R., Reese, S., Santos, J., Ingels, A., Peehl, D. AMER ASSOC CANCER RESEARCH. 2013
  • PRE-CLINICAL TRIAL OF A NEW DUAL MTOR INHIBITOR: INK128 FOR RENAL CELL CARCINOMA Ingels, A., Thong, A., Saar, M., Valta, M., Nolley, R., Santos, J., Zhao, H., Peehl, D. ELSEVIER SCIENCE INC. 2013: E65
  • Transcription Factor KLLN Inhibits Tumor Growth by AR Suppression, Induces Apoptosis by TP53/TP73 Stimulation in Prostate Carcinomas, and Correlates With Cellular Differentiation JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Wang, Y., Radhakrishnan, D., He, X., Peehl, D. M., Eng, C. 2013; 98 (3): E586-E594

    Abstract

    KLLN is a newly identified gene with unknown function and shares a bidirectional promoter with PTEN.The objective of the study was to analyze the relationship between KILLIN (KLLN) expression and prostate cancer and the potential tumor suppressive effect.We conducted an in silico analysis to compare KLLN expression in normal prostate and matched primary carcinoma tissues. We subsequently used immunohistochemistry to examine KLLN expression and association with Gleason grade and score in 109 prostatectomy samples. KLLN's tumor-suppressive effect was studied in androgen-dependent and androgen-independent cell models.Patients were diagnosed with peripheral zone prostate carcinomas without metastasis at the time of prostatectomy. Each patient's primary tumor comprised at least 2 tumoral regions with different Gleason grades.KLLN expression decreased from normal prostate tissue to primary carcinomas (P < .0001). The loss of epithelial and stromal KLLN expression is associated with poor differentiation and high Gleason scores (P < .0001), consistent with our in vitro observation that KLLN inhibits tumor cell proliferation and invasiveness. KLLN decreases prostate-specific antigen levels and suppresses androgen-mediated cell growth by inhibiting androgen receptor (AR) transcription. As an androgen receptor-regulated target, KLLN also functions as a transcriptional activator, directly promoting the expression of TP53 and TP73, with consequent elevated apoptosis, regardless of AR status.Our observations suggest that KLLN is a transcription factor directly regulating AR, TP53, and TP73 expression, with a role in prostate carcinogenesis. Loss of KLLN associates with high Gleason scores, suggesting that KLLN might be used as a potential prognostic marker for risk management and as a novel therapy target for advanced prostate carcinomas.

    View details for DOI 10.1210/jc.2012-3490

    View details for Web of Science ID 000316417200022

    View details for PubMedID 23386643

  • Anti-Oligomannose Antibodies as Potential Serum Biomarkers of Aggressive Prostate Cancer. Drug development research Wang, D., Dafik, L., Nolley, R., Huang, W., Wolfinger, R. D., Wang, L. X., Peehl, D. M. 2013; 74 (2): 65-80

    Abstract

    [Table: see text] This study bridges a carbohydrate microarray discovery and a large-scale serological validation of anti-oligomannose antibodies as novel serum biomarkers of aggressive prostate cancer (PCa). Experimentally, a Man9-cluster-specific enzyme-linked immunosorbent assay was established to enable sensitive detection of anti-Man9 antibodies in human sera. A large-cohort of men with PCa or benign prostatic hyperplasia (BPH) whose sera were banked at Stanford University was characterized using this assay. Subjects included patients with 100% Gleason grade 3 cancer (n = 84), with Gleason grades 4 and/or 5 cancer (n = 204), and BPH controls (n = 135). Radical prostatectomy Gleason grades and biochemical (PSA) recurrence served as key parameters for serum biomarker evaluation. It was found that IgGMan9 and IgMMan9 were widely present in the sera of men with BPH, as well as those with cancer. However, these antibody reactivities were significantly increased in the subjects with the largest volumes of high grade cancer. Detection of serum IgGMan9 and IgMMan9 significantly predicted the clinical outcome of PCa post-radical prostatectomy. Given these results, we suggest that IgGMan9 and IgMMan9 are novel serum biomarkers for monitoring aggressive progression of PCa. The potential of oligomannosyl antigens as targets for PCa subtyping and targeted immunotherapy is yet to be explored.

    View details for DOI 10.1002/ddr.21063

    View details for PubMedID 25152555

    View details for PubMedCentralID PMC4139012

  • Anti-Oligomannose Antibodies as Potential Serum Biomarkers of Aggressive Prostate Cancer DRUG DEVELOPMENT RESEARCH Wang, D., Dafik, L., Nolley, R., Huang, W., Wolfinger, R. D., Wang, L., Peehl, D. M. 2013; 74 (2): 65-80

    Abstract

    [Table: see text] This study bridges a carbohydrate microarray discovery and a large-scale serological validation of anti-oligomannose antibodies as novel serum biomarkers of aggressive prostate cancer (PCa). Experimentally, a Man9-cluster-specific enzyme-linked immunosorbent assay was established to enable sensitive detection of anti-Man9 antibodies in human sera. A large-cohort of men with PCa or benign prostatic hyperplasia (BPH) whose sera were banked at Stanford University was characterized using this assay. Subjects included patients with 100% Gleason grade 3 cancer (n = 84), with Gleason grades 4 and/or 5 cancer (n = 204), and BPH controls (n = 135). Radical prostatectomy Gleason grades and biochemical (PSA) recurrence served as key parameters for serum biomarker evaluation. It was found that IgGMan9 and IgMMan9 were widely present in the sera of men with BPH, as well as those with cancer. However, these antibody reactivities were significantly increased in the subjects with the largest volumes of high grade cancer. Detection of serum IgGMan9 and IgMMan9 significantly predicted the clinical outcome of PCa post-radical prostatectomy. Given these results, we suggest that IgGMan9 and IgMMan9 are novel serum biomarkers for monitoring aggressive progression of PCa. The potential of oligomannosyl antigens as targets for PCa subtyping and targeted immunotherapy is yet to be explored.

    View details for DOI 10.1002/ddr.21063

    View details for Web of Science ID 000316463700002

    View details for PubMedCentralID PMC4139012

  • Mouse model of primary human renal cell carcinoma metastasis to bone Valta, M., Zhao, H., Ingels, A., Thong, A., Nolley, R., Saar, M., Peehl, D. AMER ASSOC CANCER RESEARCH. 2013
  • Induced pluripotency of human prostatic epithelial cells. PloS one Zhao, H., Sun, N., Young, S. R., Nolley, R., Santos, J., Wu, J. C., Peehl, D. M. 2013; 8 (5)

    Abstract

    Induced pluripotent stem (iPS) cells are a valuable resource for discovery of epigenetic changes critical to cell type-specific differentiation. Although iPS cells have been generated from other terminally differentiated cells, the reprogramming of normal adult human basal prostatic epithelial (E-PZ) cells to a pluripotent state has not been reported. Here, we attempted to reprogram E-PZ cells by forced expression of Oct4, Sox2, c-Myc, and Klf4 using lentiviral vectors and obtained embryonic stem cell (ESC)-like colonies at a frequency of 0.01%. These E-PZ-iPS-like cells with normal karyotype gained expression of pluripotent genes typical of iPS cells (Tra-1-81, SSEA-3, Nanog, Sox2, and Oct4) and lost gene expression characteristic of basal prostatic epithelial cells (CK5, CK14, and p63). E-PZ-iPS-like cells demonstrated pluripotency by differentiating into ectodermal, mesodermal, and endodermal cells in vitro, although lack of teratoma formation in vivo and incomplete demethylation of pluripotency genes suggested only partial reprogramming. Importantly, E-PZ-iPS-like cells re-expressed basal epithelial cell markers (CD44, p63, MAO-A) in response to prostate-specific medium in spheroid culture. Androgen induced expression of androgen receptor (AR), and co-culture with rat urogenital sinus further induced expression of prostate-specific antigen (PSA), a hallmark of secretory cells, suggesting that E-PZ-iPS-like cells have the capacity to differentiate into prostatic basal and secretory epithelial cells. Finally, when injected into mice, E-PZ-iPS-like cells expressed basal epithelial cell markers including CD44 and p63. When co-injected with rat urogenital mesenchyme, E-PZ-iPS-like cells expressed AR and expression of p63 and CD44 was repressed. DNA methylation profiling identified epigenetic changes in key pathways and genes involved in prostatic differentiation as E-PZ-iPS-like cells converted to differentiated AR- and PSA-expressing cells. Our results suggest that iPS-like cells derived from prostatic epithelial cells are pluripotent and capable of prostatic differentiation; therefore, provide a novel model for investigating epigenetic changes involved in prostate cell lineage specification.

    View details for DOI 10.1371/journal.pone.0064503

    View details for PubMedID 23717621

    View details for PubMedCentralID PMC3661502

  • Transcript Levels of Androgen Receptor Variant AR-V1 or AR-V7 Do Not Predict Recurrence in Patients with Prostate Cancer at Indeterminate Risk for Progression JOURNAL OF UROLOGY Zhao, H., Coram, M. A., Nolley, R., Reese, S. W., Young, S. R., Peehl, D. M. 2012; 188 (6): 2158-2164

    Abstract

    AR-V7, a ligand independent splice variant of androgen receptor, may support the growth of castration resistant prostate cancer and have prognostic value. Another variant, AR-V1, interferes with AR-V7 activity. We investigated whether AR-V7 or V1 expression would predict biochemical recurrence in men at indeterminate (about 50%) risk for progression following radical prostatectomy.AR-V7 and V1 transcripts in a mixed grade cohort of 53 men in whom cancer contained 30% to 70% Gleason grade 4/5 and in a grade 3 only cohort of 52 were measured using a branched chain DNA assay. Spearman rank correlations of the transcripts, and histomorphological and clinical variables were determined. AR-V7 and V1 levels were assessed as determinants of recurrence in the mixed grade cohort by logistic regression and survival analysis. The impact of TMPRSS2-ERG gene fusion on prognosis was also evaluated.Neither AR-V7 nor V1 levels in grade 3 or 4/5 cancer in the mixed grade cohort were associated with recurrence or time to recurrence. However, AR-V7 and V1 inversely correlated with serum prostate specific antigen and positively correlated with age. The AR-V1 level in grade 3 cancer in the grade 3 only cohort was higher than in grade 3 or grade 4/5 components of mixed grade cancer. TMPRSS2-ERG fusion was not associated with AR-V7, AR-V1 or recurrence but it was associated with the percent of grade 4/5 cancer.The AR-V1 or V7 transcript level does not predict recurrence in patients with high grade prostate cancer at indeterminate risk for progression. Grade 3 cancer in mixed grade tumors may differ from 100% grade 3 cancer, at least in AR-V1 expression.

    View details for DOI 10.1016/j.juro.2012.08.014

    View details for Web of Science ID 000311581400035

    View details for PubMedID 23088973

  • Darinaparsin: Solid Tumor Hypoxic Cytotoxin and Radiosensitizer CLINICAL CANCER RESEARCH Tian, J., Zhao, H., Nolley, R., Reese, S. W., Young, S. R., Li, X., Peehl, D. M., Knox, S. J. 2012; 18 (12): 3366-3376

    Abstract

    Hypoxia is an important characteristic of the solid tumor microenvironment and constitutes a barrier for effective radiotherapy. Here, we studied the effects of darinaparsin (an arsenic cytotoxin) on survival and radiosensitivity of tumor cells in vitro under normoxia and hypoxia and in vivo using xenograft models, compared to effects on normal tissues.The cytotoxicity and radiosensitization of darinaparsin were first tested in vitro in a variety of solid tumor cell lines under both normoxia and hypoxia and compared with arsenic trioxide (ATO, an arsenical with reported cytotoxic and radiosensitizing activities on tumor cells). The effects were then tested in mouse models of xenograft tumors derived from tumor cell lines and clinical tumor specimens. The potential mechanisms of darinaparsin effects, including reactive oxygen species (ROS) generation, cellular damage, and changes in global gene expression, were also investigated.In comparison with ATO, darinaparsin had significantly higher in vitro cytotoxic and radiosensitizing activities against solid tumor cells under both normoxia and hypoxia. In vivo experiments confirmed these activities at doses that had no systemic toxicities. Importantly, darinaparsin did not radiosensitize normal bone marrow and actually radioprotected normal intestinal crypts. The darinaparsin-mediated antitumor effects under hypoxia were not dependent on ROS generation and oxidative damage, but were associated with inhibition of oncogene (RAS and MYC)-dependent gene expression.Darinaparsin has significant and preferential cytotoxic and radiosensitizing effects on solid tumors as compared with normal cells. Darinaparsin may therefore increase the therapeutic index of radiation therapy and has near term translational potential.

    View details for DOI 10.1158/1078-0432.CCR-11-3179

    View details for Web of Science ID 000307502100017

    View details for PubMedID 22535156

  • Suppression of Tak1 Promotes Prostate Tumorigenesis CANCER RESEARCH Wu, M., Shi, L., Cimic, A., Romero, L., Sui, G., Lees, C. J., Cline, J. M., Seals, D. F., Sirintrapun, J. S., McCoy, T. P., Liu, W., Kim, J. W., Hawkins, G. A., Peehl, D. M., Xu, J., Cramer, S. D. 2012; 72 (11): 2833-2843

    Abstract

    More than 30% of primary prostate cancers contain a consensus deletion of an approximately 800 kb locus on chromosome 6q15.1. The MAP3K7 gene, which encodes TGF-β activated kinase-1 (Tak1), is a putative prostate tumor suppressor gene within this region whose precise function remains obscure. In this study, we investigated the role of Tak1 in human and murine prostate cancers. In 50 well-characterized human cancer specimens, we found that Tak1 expression was progressively lost with increasing Gleason grade, both within each cancer and across all cancers. In murine prostate stem cells and Tak1-deficient prostatic epithelial cells, Tak1 loss increased proliferation, migration, and invasion. When prostate stem cells attenuated for Tak1 were engrafted with fetal urogenital mesenchyme, the histopathology of the grafts reflected the natural history of prostate cancer leading from prostatic intraepithelial neoplasia to invasive carcinoma. In the grafts containing Tak1-suppressed prostate stem cells, p38 and c-jun-NH(2)-kinase activity was attenuated and proliferation was increased. Together, our findings functionally validate the proposed tumor suppressor role of Tak1 in prostate cancer.

    View details for DOI 10.1158/0008-5472.CAN-11-2724

    View details for Web of Science ID 000307348000016

    View details for PubMedID 22467172

    View details for PubMedCentralID PMC3654674

  • Microfluidic integration for automated targeted proteomic assays PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Hughes, A. J., Lin, R. K., Peehl, D. M., Herr, A. E. 2012; 109 (16): 5972-5977

    Abstract

    A dearth of protein isoform-based clinical diagnostics currently hinders advances in personalized medicine. A well-organized protein biomarker validation process that includes facile measurement of protein isoforms would accelerate development of effective protein-based diagnostics. Toward scalable protein isoform analysis, we introduce a microfluidic "single-channel, multistage" immunoblotting strategy. The multistep assay performs all immunoblotting steps: separation, immobilization of resolved proteins, antibody probing of immobilized proteins, and all interim wash steps. Programmable, low-dispersion electrophoretic transport obviates the need for pumps and valves. A three-dimensional bulk photoreactive hydrogel eliminates manual blotting. In addition to simplified operation and interfacing, directed electrophoretic transport through our 3D nanoporous reactive hydrogel yields superior performance over the state-of-the-art in enhanced capture efficiency (on par with membrane electroblotting) and sparing consumption of reagents (ca. 1 ng antibody), as supported by empirical and by scaling analyses. We apply our fully integrated microfluidic assay to protein measurements of endogenous prostate specific antigen isoforms in (i) minimally processed human prostate cancer cell lysate (1.1 pg limit of detection) and (ii) crude sera from metastatic prostate cancer patients. The single-instrument functionality establishes a scalable microfluidic framework for high-throughput targeted proteomics, as is relevant to personalized medicine through robust protein biomarker verification, systematic characterization of new antibody probes for functional proteomics, and, more broadly, to characterization of human biospecimen repositories.

    View details for DOI 10.1073/pnas.1108617109

    View details for Web of Science ID 000303246100022

    View details for PubMedID 22474344

    View details for PubMedCentralID PMC3341062

  • Integrated Analysis Reveals Critical Genomic Regions in Prostate Tumor Microenvironment Associated with Clinicopathologic Phenotypes CLINICAL CANCER RESEARCH Ashida, S., Orloff, M. S., Bebek, G., Zhang, L., Zheng, P., Peehl, D. M., Eng, C. 2012; 18 (6): 1578-1587

    Abstract

    Recent studies suggest that tumor microenvironment (stroma) is important in carcinogenesis and progression. We sought to integrate global genomic structural and expressional alterations in prostate cancer epithelium and stroma and their association with clinicopathologic features.We conducted a genome-wide LOH/allelic imbalance (AI) scan of DNA from epithelium and stroma of 116 prostate cancers. LOH/AI hot or cold spots were defined as the markers with significantly higher or lower LOH/AI frequencies compared with the average frequency for markers along the same chromosome. These data were then integrated with publicly available transcriptome data sets and our experimentally derived data. Immunohistochemistry on an independent series was used for validation.Overall, we identified 43 LOH/AI hot/cold spots, 17 in epithelium and stroma (P < 0.001), 18 only in epithelium (P < 0.001), and eight only in stroma (P < 0.001). Hierarchical clustering of expression data supervised by genes within LOH/AI hot/cold spots in both epithelium and stroma accurately separated samples into normal epithelium, primary cancer, and metastatic cancer groups, which could not be achieved with data from only epithelium. Importantly, our experimental expression data of the genes within the LOH/AI hot/cold spots in stroma accurately clustered normal stroma from cancer stroma. We also identified 15 LOH/AI markers that were associated with Gleason score, which were validated functionally in each compartment by transcriptome data. Independent immunohistochemical validation of STIM2 within a stromal significant LOH marker (identified as associated with Gleason grade) confirmed its downregulation in the transition from moderate to high Gleason grade.Compartment-specific genomic and transcriptomic alterations accurately distinguish clinical and pathologic outcomes, suggesting new biomarkers for prognosis and targeted therapeutics.

    View details for DOI 10.1158/1078-0432.CCR-11-2535

    View details for Web of Science ID 000301672400015

    View details for PubMedID 22275508

  • Cell surface glycoproteomic analysis of prostate cancer-derived PC-3 cells BIOORGANIC & MEDICINAL CHEMISTRY LETTERS Hubbard, S. C., Boyce, M., McVaugh, C. T., Peehl, D. M., Bertozzi, C. R. 2011; 21 (17): 4945-4950

    Abstract

    Most clinically approved biomarkers of cancer are glycoproteins, and those residing on the cell surface are of particular interest in biotherapeutics. We report a method for selective labeling, affinity enrichment, and identification of cell-surface glycoproteins. PC-3 cells and primary human prostate cancer tissue were treated with peracetylated N-azidoacetylgalactosamine, resulting in metabolic labeling of cell surface glycans with the azidosugar. We used mass spectrometry to identify over 70 cell surface glycoproteins and biochemically validated CD146 and integrin beta-4, both of which are known to promote metastatic behavior. These results establish cell-surface glycoproteomics as an effective technique for discovery of cancer biomarkers.

    View details for DOI 10.1016/j.bmcl.2011.05.045

    View details for Web of Science ID 000293884100002

    View details for PubMedID 21798741

    View details for PubMedCentralID PMC3341932

  • DNA methylation profiling reveals novel biomarkers and important roles for DNA methyltransferases in prostate cancer GENOME RESEARCH Kobayashi, Y., Absher, D. M., Gulzar, Z. G., Young, S. R., McKenney, J. K., Peehl, D. M., Brooks, J. D., Myers, R. M., Sherlock, G. 2011; 21 (7): 1017-1027

    Abstract

    Candidate gene-based studies have identified a handful of aberrant CpG DNA methylation events in prostate cancer. However, DNA methylation profiles have not been compared on a large scale between prostate tumor and normal prostate, and the mechanisms behind these alterations are unknown. In this study, we quantitatively profiled 95 primary prostate tumors and 86 benign adjacent prostate tissue samples for their DNA methylation levels at 26,333 CpGs representing 14,104 gene promoters by using the Illumina HumanMethylation27 platform. A 2-class Significance Analysis of this data set revealed 5912 CpG sites with increased DNA methylation and 2151 CpG sites with decreased DNA methylation in tumors (FDR < 0.8%). Prediction Analysis of this data set identified 87 CpGs that are the most predictive diagnostic methylation biomarkers of prostate cancer. By integrating available clinical follow-up data, we also identified 69 prognostic DNA methylation alterations that correlate with biochemical recurrence of the tumor. To identify the mechanisms responsible for these genome-wide DNA methylation alterations, we measured the gene expression levels of several DNA methyltransferases (DNMTs) and their interacting proteins by TaqMan qPCR and observed increased expression of DNMT3A2, DNMT3B, and EZH2 in tumors. Subsequent transient transfection assays in cultured primary prostate cells revealed that DNMT3B1 and DNMT3B2 overexpression resulted in increased methylation of a substantial subset of CpG sites that showed tumor-specific increased methylation.

    View details for DOI 10.1101/gr.119487.110

    View details for Web of Science ID 000292298000003

    View details for PubMedID 21521786

    View details for PubMedCentralID PMC3129245

  • The Alternative Splice Variant of Protein Tyrosine Kinase 6 Negatively Regulates Growth and Enhances PTK6-Mediated Inhibition of beta-Catenin PLOS ONE Brauer, P. M., Zheng, Y., Evans, M. D., Dominguez-Brauer, C., Peehl, D. M., Tyner, A. L. 2011; 6 (3)

    Abstract

    Protein tyrosine kinase 6 (PTK6), also called breast tumor kinase (BRK), is expressed in epithelial cells of various tissues including the prostate. Previously it was shown that PTK6 is localized to epithelial cell nuclei in normal prostate, but becomes cytoplasmic in human prostate tumors. PTK6 is also primarily cytoplasmic in the PC3 prostate adenocarcinoma cell line. Sequencing revealed expression of wild type full-length PTK6 transcripts in addition to an alternative transcript lacking exon 2 in PC3 cells. The alternative transcript encodes a 134 amino acid protein, referred to here as ALT-PTK6, which shares the first 77 amino acid residues including the SH3 domain with full length PTK6. RT-PCR was used to show that ALT-PTK6 is coexpressed with full length PTK6 in established human prostate and colon cell lines, as well as in primary cell lines derived from human prostate tissue and tumors. Although interaction between full-length PTK6 and ALT-PTK6 was not detected, ALT-PTK6 associates with the known PTK6 substrates Sam68 and β-catenin in GST pull-down assays. Coexpression of PTK6 and ALT-PTK6 led to suppression of PTK6 activity and reduced association of PTK6 with tyrosine phosphorylated proteins. While ALT-PTK6 alone did not influence β-catenin/TCF transcriptional activity in a luciferase reporter assay, it enhanced PTK6-mediated inhibition of β-catenin/TCF transcription by promoting PTK6 nuclear functions. Ectopic expression of ALT-PTK6 led to reduced expression of the β-catenin/TCF targets Cyclin D1 and c-Myc in PC3 cells. Expression of tetracycline-inducible ALT-PTK6 blocked the proliferation and colony formation of PC3 cells. Our findings suggest that ALT-PTK6 is able to negatively regulate growth and modulate PTK6 activity, protein-protein associations and/or subcellular localization. Fully understanding functions of ALT-PTK6 and its impact on PTK6 signaling will be critical for development of therapeutic strategies that target PTK6 in cancer.

    View details for DOI 10.1371/journal.pone.0014789

    View details for Web of Science ID 000289055700002

    View details for PubMedID 21479203

    View details for PubMedCentralID PMC3068133

  • Serum Mac-2BP Does Not Distinguish Men With High Grade, Large Volume Prostate Cancer From Men With Benign Prostatic Hyperplasia PROSTATE Peehl, D. M., Chen, Z., Nolley, R. 2011; 71 (1): 26-31

    Abstract

    Mac-2 binding protein (Mac-2BP) is a secreted protein that has been used as a serum prognostic marker for several types of cancers. A previous study showed that serum Mac-2BP was significantly higher (∼2-fold) in men with prostate cancer compared to healthy men. We investigated whether serum Mac-2BP could distinguish men with high grade, large volume prostate cancer from men with benign prostatic hyperplasia (BPH).A commercially available ELISA kit was used to measure Mac-2BP in paired pre- and post-prostatectomy sera from 10 men with high grade, large volume prostate cancer, in pre-operative sera from 50 untreated men with high grade, large volume prostate cancer, and in sera from 50 men with clinical symptoms of BPH and biopsy-negative for prostate cancer. Results were analyzed by Student's t-test and receiver operating characteristic (ROC) curves.Levels of Mac-2BP did not decrease in post-prostatectomy sera, and Mac-2BP values were not significantly different in the sera of men with prostate cancer versus those with BPH.Serum Mac-2BP does not appear to originate in the prostate and it is unlikely that Mac-2BP can be used for the differential diagnosis of prostate cancer versus BPH.

    View details for DOI 10.1002/pros.21218

    View details for Web of Science ID 000285849000003

    View details for PubMedID 20583127

  • Stiffness mapping prostate biopsy samples using a tactile sensor 33rd Annual International Conference of the IEEE Engineering-in-Medicine-and-Biology-Society (EMBS) Peng, Q., Omata, S., Peehl, D. M., Constantinou, C. E. IEEE. 2011: 8515–8518

    Abstract

    Previous studies have demonstrated that the stiffness of cancerous cells reflects their pathological stage and progression rates, with increased cancerous cell stiffness associated with increased aggressiveness. Therefore, the elasticity of the cancerous cells has the potential to be used as an indicator of the cancer's aggressiveness. However, the sensitivity and resolution of current palpation and imaging techniques are not sufficient to detect small cancerous tissues. In previous studies, we developed a tactile-based device to map with high resolution the stiffness of a tissue section. The purpose of this study is to evaluate this device using different tissues (BPH, Cancer and PZ) collected from human prostates. The preliminary results show that the tactile device is sensitive enough to tell the differences of the stiffness of different tissues. The results also disclosed the factors (humidity, temperature and tissue degradation) which could dramatically affect the results of stiffness mapping. The tactile technology described in this paper has the potential to help disclose the underlying mechanical mechanisms that lead to increased stiffness in prostate tumors.

    View details for Web of Science ID 000298810006159

    View details for PubMedID 22256325

  • Anti-tumor and radiosensitization activities of the iron chelator HDp44mT are mediated by effects on intracellular redox status CANCER LETTERS Tian, J., Peehl, D. M., Zheng, W., Knox, S. J. 2010; 298 (2): 231-237

    Abstract

    A novel iron chelator, HDp44mT, has been reported to have potent anti-proliferative effects on cancer cells; however, the underlying mechanism of action is not well understood. In this study, we characterized the cytotoxic effect of HDp44mT in a chemo- and radio-resistant cell line (PC-3) of prostatic cancer origin. The activity of HDp44mT at nM concentrations was dependent on the intracellular GSH and atmospheric O(2) concentration, rather than iron deprivation. HDp44mT also radiosensitized PC-3 cells in a GSH-dependent manner. Interestingly, this radiosensitizing effect was observed under aerobic and, to a larger extent, hypoxic conditions, suggesting its potential utility as a radiosensitizer for some radioresistant tumors.

    View details for DOI 10.1016/j.canlet.2010.07.010

    View details for Web of Science ID 000284300000011

    View details for PubMedID 20678860

  • Targeting monoamine oxidase A in advanced prostate cancer JOURNAL OF CANCER RESEARCH AND CLINICAL ONCOLOGY Flamand, V., Zhao, H., Peehl, D. M. 2010; 136 (11): 1761-1771

    Abstract

    Inhibitors of monoamine oxidase A (MAOA), a mitochondrial enzyme that degrades neurotransmitters including serotonin and norepinephrine, are commonly used to treat neurological conditions including depression. Recently, we and others identified high expression of MAOA in normal basal prostatic epithelium and high-grade primary prostate cancer (PCa). In contrast, MAOA is low in normal secretory prostatic epithelium and low-grade PCa. An irreversible inhibitor of MAOA, clorgyline, induced secretory differentiation in primary cultures of normal basal epithelial cells and high-grade PCa. Furthermore, clorgyline inhibited several oncogenic pathways in PCa cells, suggesting clinical value of MAOA inhibitors as a pro-differentiation and anti-oncogenic therapy for high-risk PCa. Here, we extended our studies to a model of advanced PCa, VCaP cells, which were derived from castration-resistant metastatic PCa and express a high level of MAOA.Growth of VCaP cells in the presence or absence of clorgyline was evaluated in vitro and in vivo. Gene expression changes in response to clorgyline were determined by microarray and validated by quantitative real-time polymerase chain reaction.Treatment with clorgyline in vitro inhibited growth and altered the transcriptional pattern of VCaP cells in a manner consistent with the pro-differentiation and anti-oncogenic effects seen in treated primary PCa cells. Src, beta-catenin, and MAPK oncogenic pathways, implicated in androgen-independent growth and metastasis, were significantly downregulated. Clorgyline treatment of mice bearing VCaP xenografts slowed tumor growth and induced transcriptome changes similar to those noted in vitro.Our results support the possibility that anti-depressant drugs that target MAOA might find a new application in treating PCa.

    View details for DOI 10.1007/s00432-010-0835-6

    View details for Web of Science ID 000282183700016

    View details for PubMedID 20204405

    View details for PubMedCentralID PMC2945406

  • Metalloporphyrin Synergizes with Ascorbic Acid to Inhibit Cancer Cell Growth Through Fenton Chemistry CANCER BIOTHERAPY AND RADIOPHARMACEUTICALS Tian, J., Peehl, D. M., Knox, S. J. 2010; 25 (4): 439-448

    Abstract

    Ascorbic acid (AA) has been reported to inhibit tumor cell growth through the generation of extracellular hydrogen peroxide (H(2)O(2)). However, the clinical utility of AA has been limited by relatively low potency and in vivo efficacy. This study reports that the metalloporphyrin, Mn(III) tetrakis(N-methylpyridinium-2-yl)porphyrin(5+) (MnTMPyP), has a potent synergistic cytotoxic effect when combined with AA in a variety of cancer cell lines. In the presence of MnTMPyP, the concentration of AA required to inhibit cancer cell growth was markedly reduced. In vitro (cell-free) experiments demonstrated that AA alone enhanced the Fenton reaction that produces cytotoxic hydroxyl radical (HO(*)); however, this reaction was limited by the low rate by which AA generates H(2)O(2) (Fenton reaction substrate) from O(2). MnTMPyP catalyzed H(2)O(2) generation through the AA-facilitated Mn(II <--> III)TMPyP redox cycle and thereby markedly potentiated the Fenton reaction. Accordingly, MnTMPyP and AA resulted in increased cellular levels of H(2)O(2) and HO(*) in cancer cells, which mediate the synergistic cytotoxicity of this combined treatment. This effect was inhibited by cellular enzymes that metabolize H(2)O(2), such as catalase and glutathione peroxidase, suggesting that selective killing of cancer cells deficient in such enzymes can be achieved in vivo.

    View details for DOI 10.1089/cbr.2009.0756

    View details for Web of Science ID 000281255100008

    View details for PubMedID 20735206

  • Tissue Slice Grafts An in Vivo Model of Human Prostate Androgen Signaling AMERICAN JOURNAL OF PATHOLOGY Zhao, H., Nolley, R., Chen, Z., Peehl, D. M. 2010; 177 (1): 229-239

    Abstract

    We developed a tissue slice graft (TSG) model by implanting thin, precision-cut tissue slices derived from fresh primary prostatic adenocarcinomas under the renal capsule of immunodeficient mice. This new in vivo model not only allows analysis of approximately all of the cell types present in prostate cancer within an intact tissue microenvironment, but also provides a more accurate assessment of the effects of interventions when tissues from the same specimen with similar cell composition and histology are used as control and experimental samples. The thinness of the slices ensures that sufficient samples can be obtained for large experiments as well as permits optimal exchange of nutrients, oxygen, and drugs between the grafted tissue and the host. Both benign and cancer tissues displayed characteristic histology and expression of cell-type specific markers for up to 3 months. Moreover, androgen-regulated protein expression diminished in TSGs after androgen ablation of the host and was restored after androgen repletion. Finally, many normal secretory epithelial cells and cancer cells in TSGs remained viable 2 months after androgen ablation, consistent with similar observations in postprostatectomy specimens following neoadjuvant androgen ablation. Among these were putative Nkx3.1(+) stem cells. Our novel TSG model has the appropriate characteristics to serve as a useful tool to model all stages of disease, including normal tissue, premalignant lesions, well-differentiated cancer, and poorly differentiated cancer.

    View details for DOI 10.2353/ajpath.2010.090821

    View details for Web of Science ID 000279805100025

    View details for PubMedID 20472887

    View details for PubMedCentralID PMC2893666

  • Methods for Metabolic Evaluation of Prostate Cancer Cells Using Proton and C-13 HR-MAS Spectroscopy and [3-C-13] Pyruvate as a Metabolic Substrate MAGNETIC RESONANCE IN MEDICINE Levin, Y. S., Albers, M. J., Butler, T. N., Spielman, D., Peehl, D. M., Kurhanewicz, J. 2009; 62 (5): 1091-1098

    Abstract

    Prostate cancer has been shown to undergo unique metabolic changes associated with neoplastic transformation, with associated changes in citrate, alanine, and lactate concentrations. (13)C high resolution-magic angle spinning (HR-MAS) spectroscopy provides an opportunity to simultaneously investigate the metabolic pathways implicated in these changes by using (13)C-labeled substrates as metabolic probes. In this work, a method to reproducibly interrogate metabolism in prostate cancer cells in primary culture was developed using HR-MAS spectroscopy. Optimization of cell culture protocols, labeling parameters, harvesting, storage, and transfer was performed. Using [3-(13)C] pyruvate as a metabolic probe, (1)H and (13)C HR-MAS spectroscopy was used to quantify the net amount and fractional enrichment of several labeled metabolites that evolved in multiple cell samples from each of five different prostate cancers. Average enrichment across all cancers was 32.4 +/- 5.4% for [3-(13)C] alanine, 24.5 +/- 5.4% for [4-(13)C] glutamate, 9.1 +/- 2.5% for [3-(13)C] glutamate, 25.2 +/- 5.7% for [3-(13)C] aspartate, and 4.2 +/- 1.0% for [3-(13)C] lactate. Cell samples from the same parent population demonstrated reproducible fractional enrichments of alanine, glutamate, and aspartate to within 12%, 10%, and 10%, respectively. Furthermore, the cells produced a significant amount of [4-(13)C] glutamate, which supports the bioenergetic theory for prostate cancer. These methods will allow further characterization of metabolic properties of prostate cancer cells in the future. Magn Reson Med, 2009. (c) 2009 Wiley-Liss, Inc.

    View details for DOI 10.1002/mrm.22120

    View details for Web of Science ID 000271431200001

    View details for PubMedID 19780158

    View details for PubMedCentralID PMC2783184

  • Anti-oncogenic and pro-differentiation effects of clorgyline, a monoamine oxidase A inhibitor, on high grade prostate cancer cells BMC MEDICAL GENOMICS Zhao, H., Flamand, V., Peehl, D. M. 2009; 2

    Abstract

    Monoamine oxidase A (MAO-A), a mitochondrial enzyme that degrades monoamines including neurotransmitters, is highly expressed in basal cells of the normal human prostatic epithelium and in poorly differentiated (Gleason grades 4 and 5), aggressive prostate cancer (PCa). Clorgyline, an MAO-A inhibitor, induces secretory differentiation of normal prostate cells. We examined the effects of clorgyline on the transcriptional program of epithelial cells cultured from high grade PCa (E-CA).We systematically assessed gene expression changes induced by clorgyline in E-CA cells using high-density oligonucleotide microarrays. Genes differentially expressed in treated and control cells were identified by Significance Analysis of Microarrays. Expression of genes of interest was validated by quantitative real-time polymerase chain reaction.The expression of 156 genes was significantly increased by clorgyline at all time points over the time course of 6 - 96 hr identified by Significance Analysis of Microarrays (SAM). The list is enriched with genes repressed in 7 of 12 oncogenic pathway signatures compiled from the literature. In addition, genes downregulated >or= 2-fold by clorgyline were significantly enriched with those upregulated by key oncogenes including beta-catenin and ERBB2, indicating an anti-oncogenic effect of clorgyline. Another striking effect of clorgyline was the induction of androgen receptor (AR) and classic AR target genes such as prostate-specific antigen together with other secretory epithelial cell-specific genes, suggesting that clorgyline promotes differentiation of cancer cells. Moreover, clorgyline downregulated EZH2, a critical component of the Polycomb Group (PcG) complex that represses the expression of differentiation-related genes. Indeed, many genes in the PcG repression signature that predicts PCa outcome were upregulated by clorgyline, suggesting that the differentiation-promoting effect of clorgyline may be mediated by its downregulation of EZH2.Our results suggest that inhibitors of MAO-A, already in clinical use to treat depression, may have potential application as therapeutic PCa drugs by inhibiting oncogenic pathway activity and promoting differentiation.

    View details for DOI 10.1186/1755-8794-2-55

    View details for Web of Science ID 000272814900001

    View details for PubMedID 19691856

    View details for PubMedCentralID PMC2736984

  • Tumor-Promoting Phenotype of CD90(hi) Prostate Cancer-Associated Fibroblasts PROSTATE Zhao, H., Peehl, D. M. 2009; 69 (9): 991-1000

    Abstract

    Cancer-associated stroma contributes to the malignant behavior of adenocarcinomas of the prostate and other organs. CD90 is a marker of mesenchymal stem cells (MSCs) and its expression is higher in prostate cancer stroma compared to normal tissue. Cultured prostate cancer-associated fibroblasts (CAFs) expressing high versus low levels of CD90 were analyzed for an MSC-like or tumor-promoting phenotype.CD90(hi) and CD90(lo) cells were collected by fluorescence-activated cell sorting (FACS). Expression of genes associated with MSCs and/or tumor-promoting activities was measured by quantitative polymerase chain reaction (qPCR). Effects of stromal cell co-culture or conditioned media were tested on BPH-1 epithelial cells.The pattern of gene expression did not support the hypothesis that CD90(hi) cells were MSCs. However, CD90(hi) cells expressed higher levels of many genes associated with tumor promotion, including cytokines, angiogenic factors, hedgehog signaling components, and transforming growth factor (TGF)-beta. Co-culture or conditioned medium from CD90(hi) cells increased CXCR4 expression in BPH-1 cells, at least in part due to TGF-beta, and protected BPH-1 cells from apoptosis.Our results suggest that the elevated expression of CD90 previously observed in the cancer-associated stroma of the human prostate is biologically significant. Although our results do not support the idea that CD90(hi) cells cultured from the cancer stroma are MSCs, our findings suggest that the phenotype of these cells is more tumor-promoting than that of cells expressing low CD90.

    View details for DOI 10.1002/pros.20946

    View details for Web of Science ID 000266470700010

    View details for PubMedID 19267366

    View details for PubMedCentralID PMC2736596

  • Inhibition of prostaglandin synthesis and actions by genistein in human prostate cancer cells and by soy isoflavones in prostate cancer patients INTERNATIONAL JOURNAL OF CANCER Swami, S., Krishnan, A. V., Moreno, J., Bhattacharya, R. S., Gardner, C., Brooks, J. D., Peehl, D. M., Feldman, D. 2009; 124 (9): 2050-2059

    Abstract

    Soy and its constituent isoflavone genistein inhibit the development and progression of prostate cancer (PCa). Our study in both cultured cells and PCa patients reveals a novel pathway for the actions of genistein, namely the inhibition of the synthesis and biological actions of prostaglandins (PGs), known stimulators of PCa growth. In the cell culture experiments, genistein decreased cyclooxygenase-2 (COX-2) mRNA and protein expression in both human PCa cell lines (LNCaP and PC-3) and primary prostate epithelial cells and increased 15-hydroxyprostaglandin dehydrogenase (15-PGDH) mRNA levels in primary prostate cells. As a result genistein significantly reduced the secretion of PGE(2) by these cells. EP4 and FP PG receptor mRNA were also reduced by genistein, providing an additional mechanism for the suppression of PG biological effects. Further, the growth stimulatory effects of both exogenous PGs and endogenous PGs derived from precursor arachidonic acid were attenuated by genistein. We also performed a pilot randomised double blind clinical study in which placebo or soy isoflavone supplements were given to PCa patients in the neo-adjuvant setting for 2 weeks before prostatectomy. Gene expression changes were measured in the prostatectomy specimens. In PCa patients ingesting isoflavones, we observed significant decreases in prostate COX-2 mRNA and increases in p21 mRNA. There were significant correlations between COX-2 mRNA suppression, p21 mRNA stimulation and serum isoflavone levels. We propose that the inhibition of the PG pathway contributes to the beneficial effect of soy isoflavones in PCa chemoprevention and/or treatment.

    View details for DOI 10.1002/ijc.24161

    View details for Web of Science ID 000264647600007

    View details for PubMedID 19127598

  • The Elevated Expression of a Mismatch Repair Protein Is a Predictor for Biochemical Recurrence After Radical Prostatectomy CANCER EPIDEMIOLOGY BIOMARKERS & PREVENTION Norris, A. M., Gentry, M., Peehl, D. M., D'Agostino, R., Scarpinato, K. D. 2009; 18 (1): 57–64

    Abstract

    The inability to predict clinical outcome of prostate cancer is a major impediment to effective treatment decisions and patient counseling. New markers of recurrence are needed to improve the accuracy of risk assessment and treatment of prostate cancer. Our previous studies identified a mismatch repair protein, PMS2, to be elevated in prostate cancer; here, we investigate the prognostic potential of this marker. We hypothesized that the elevation of PMS2 would correlate with disease outcome.Retrospective quantitative immunohistochemistry was done to measure PMS2 in high-grade cancers of 166 men treated by radical prostatectomy with a biochemical recurrence rate of 56%. Associations between PMS2 levels, pathologic variables, and biochemical recurrence over time were determined.The mean level of PMS2 protein was consistently higher in both cancer-associated benign epithelium and cancer cells of patients who recurred, compared with nonrecurrent patients. PMS2 was an independent predictor of time-to-recurrence in Cox multivariate analyses and significantly stratified patients based on outcome. PMS2 was able to improve the sensitivity of total percent Gleason 4/5 as a risk factor for recurrence in this cohort.PMS2 protein levels were shown to be a predictor of time-to-recurrence after surgery. This study is the first to document that the elevation of a mismatch repair protein negatively correlates with prognosis and has implications in patient diagnosis and molecular profiling.

    View details for DOI 10.1158/1055-9965.EPI-08-0377

    View details for Web of Science ID 000262424200008

    View details for PubMedID 19124481

    View details for PubMedCentralID PMC2701238

  • The Significance of Monoamine Oxidase-A Expression in High Grade Prostate Cancer JOURNAL OF UROLOGY Peehl, D. M., Coram, M., Khine, H., Reese, S., Nolley, R., Zhao, H. 2008; 180 (5): 2206-2211

    Abstract

    Gleason grade 4/5 prostate cancer is a determinant for recurrence following radical prostatectomy. Monoamine oxidase-A is over expressed in grade 4/5 compared to grade 3 cancer. Monoamine oxidase-A is also expressed by normal basal cells and in vitro studies suggest that its function is to repress secretory differentiation. Therefore, monoamine oxidase-A in grade 4/5 cancer might reflect dedifferentiation to a basal cell-like phenotype. We investigated whether monoamine oxidase-A expression correlates with another basal cell protein, CD44, in high grade cancer and whether either is associated with an aggressive phenotype.A total of 133 grade 4/5 archival cancers from a cohort previously used to evaluate the prognostic significance of histomorphological variables were scored for monoamine oxidase-A and CD44 immunohistochemical labeling. Spearman rank correlations of the proteins, and histomorphological and clinical variables were determined. The univariate and multivariate value of each variable as a determinant of biochemical recurrence was assessed by logistic regression.Monoamine oxidase-A expression correlated with CD44. Neither was prognostic for biochemical recurrence. However, monoamine oxidase-A expression positively correlated with preoperative serum prostate specific antigen and the percent of grade 4/5 cancer.Concurrent expression of monoamine oxidase-A and CD44 suggests that grade 4/5 cancer may be basal cell-like in nature, despite the absence of other classic basal cell biomarkers such as cytokeratins 5 and 14, and p63. The correlation of monoamine oxidase-A expression with prostate specific antigen and the percent of grade 4/5 cancer suggests that monoamine oxidase-A may contribute to growth of high grade cancer and that antidepressant drugs that target monoamine oxidase-A may have applications in treating prostate cancer.

    View details for DOI 10.1016/j.juro.2008.07.019

    View details for Web of Science ID 000260102000083

    View details for PubMedID 18804811

    View details for PubMedCentralID PMC2743604

  • Inhibition of monoamine oxidase A promotes secretory differentiation in basal prostatic epithelial cells DIFFERENTIATION Zhao, H., Nolley, R., Chen, Z., Reese, S. W., Peehl, D. M. 2008; 76 (7): 820-830

    Abstract

    Monoamine oxidase A (MAO-A) expression is associated with high-grade prostate cancer. Immunohistochemistry showed that MAO-A is also expressed in the basal epithelial cells of normal prostate glands. Using cultured primary prostatic epithelial cells as a model, we showed that MAO-A prevents basal epithelial cells from differentiating into secretory cells. Under differentiation-promoting conditions, clorgyline, an irreversible MAO-A inhibitor, induced secretory cell-like morphology and repressed expression of cytokeratin 14, a basal cell marker. More importantly, clorgyline induced mRNA and protein expression of androgen receptor (AR), a hallmark of secretory epithelial cells. In clorgyline-treated cells, androgen induced luciferase activity controlled by the promoter of prostate-specific antigen, an AR target gene, in a dose-dependent manner. This activity was blocked by the AR antagonist Casodex, showing that AR is functional. In turn, androgen decreased MAO-A expression in clorgyline-treated, secretory-like cells. Our results demonstrated that cultured basal epithelial cells have the potential to differentiate into secretory cells, and that inhibition of MAO-A is a key factor in promoting this process. Increased expression of MAO-A in high-grade prostate cancer may be an important contributor to its de-differentiated phenotype, raising the possibility that MAO-A inhibition may restore differentiation and reverse the aggressive behavior of high-grade cancer.

    View details for DOI 10.1111/j.1432-0436.2007.00263.x

    View details for Web of Science ID 000258581400007

    View details for PubMedID 18248494

    View details for PubMedCentralID PMC2760409

  • Association of prostate-specific antigen promoter genotype with clinical and histopathologic features of prostate cancer CANCER EPIDEMIOLOGY BIOMARKERS & PREVENTION Cramer, S. D., Sun, J., Zheng, S. L., Xu, J., Peehl, D. M. 2008; 17 (9): 2451-2457

    Abstract

    The serum test for the secreted protease prostate-specific antigen (PSA) is the most widely used screening tool for prostate cancer. The PSA gene contains multiple functional and nonfunctional single nucleotide polymorphisms (SNP) in its promoter. We showed previously that the rs925013 G/A SNP, but not the rs266882 G/A SNP, was significantly associated with serum PSA in healthy men. In this study, we evaluated the association of the PSA promoter genotype with clinical data in a cohort of 1,224 men with prostate cancer. Previous work with a subset of this cohort has shown that percent high-grade (Gleason grades 4 and 5) cancer was the strongest predictor of biochemical recurrence (PSA relapse). We found a statistically significant association (P < 0.05) of the rs925013 SNP with several clinical and histomorphologic variables. The G allele was associated with higher serum PSA at diagnosis, higher percent Gleason grade 3 cancer, and lower percent high-grade and Gleason grade 4 cancer. The rs266882 SNP was modestly associated with PSA at diagnosis in a dominant model but was not associated with cancer grade. Neither SNP was associated with biochemical recurrence. The statistically significant predictors of biochemical recurrence were tumor location in the peripheral zone [odds ratio (OR), 10.71; 95% confidence interval (95% CI), 3.15-36.49], presence of any Gleason grade 4/5 cancer (OR, 4.26; 95% CI, 1.30-14.00), presence of any intraductal cancer (OR, 1.03; 95% CI, 1.00-1.04), and serum PSA at diagnosis (OR, 2.04; 95% CI, 1.50-2.77).

    View details for DOI 10.1158/1055-9965.EPI-08-0374

    View details for Web of Science ID 000259211400038

    View details for PubMedID 18768516

    View details for PubMedCentralID PMC4114034

  • Centrosomal PKC beta II and pericentrin are critical for human prostate cancer growth and angiogenesis CANCER RESEARCH Kim, J., Choi, Y., Vallentin, A., Hunrichs, B. S., Hellerstein, M. K., Peehl, D. M., Mochly-Rosen, D. 2008; 68 (16): 6831-6839

    Abstract

    Angiogenesis is critical in the progression of prostate cancer. However, the interplay between the proliferation kinetics of tumor endothelial cells (angiogenesis) and tumor cells has not been investigated. Also, protein kinase C (PKC) regulates various aspects of tumor cell growth, but its role in prostate cancer has not been investigated in detail. Here, we found that the proliferation rates of endothelial and tumor cells oscillate asynchronously during the growth of human prostate cancer xenografts. Furthermore, our analyses suggest that PKCbetaII was activated during increased angiogenesis and that PKCbetaII plays a key role in the proliferation of endothelial cells and tumor cells in human prostate cancer; treatment with a PKCbetaII-selective inhibitor, betaIIV5-3, reduced angiogenesis and tumor cell proliferation. We also find a unique effect of PKCbetaII inhibition on normalizing pericentrin (a protein regulating cytokinesis), especially in endothelial cells as well as in tumor cells. PKCbetaII inhibition reduced the level and mislocalization of pericentrin and normalized microtubule organization in the tumor endothelial cells. Although pericentrin has been known to be up-regulated in epithelial cells of prostate cancers, its level in tumor endothelium has not been studied in detail. We found that pericentrin is up-regulated in human tumor endothelium compared with endothelium adjacent to normal glands in tissues from prostate cancer patients. Our results suggest that a PKCbetaII inhibitor such as betaIIV5-3 may be used to reduce prostate cancer growth by targeting both angiogenesis and tumor cell growth.

    View details for DOI 10.1158/0008-5472.CAN-07-6195

    View details for PubMedID 18701509

  • Topology of NGEP, a prostate-specific cell: Cell junction protein widely expressed in many cancers of different grade level CANCER RESEARCH Das, S., Hahn, Y., Walker, D. A., Nagata, S., Willingham, M. C., Peehl, D. M., Bera, T. K., Lee, B., Pastan, I. 2008; 68 (15): 6306-6312

    Abstract

    New gene expressed in prostate (NGEP) is a prostate-specific polytopic membrane protein found at high concentrations at cell:cell contact regions. To determine if NGEP is a useful target for antibody-based therapy of prostate cancer, we performed an immunohistochemical analysis of 126 human prostate carcinoma samples using polyclonal anti-NGEP sera and found that 91% of the cancers express NGEP protein. To elucidate the topology of NGEP and guide the development of monoclonal antibodies (mAb) reacting with the extracellular regions of NGEP, a hemagglutinin epitope tag was inserted at several positions within the NGEP sequence. The tagged proteins were expressed in 293T cells and locations of the tags were determined by immunofluorescence in intact or permeabilized cells. The results indicate that NGEP contains eight transmembrane domains with both the NH(2) and COOH termini of NGEP located inside the cell. We produced mAb to three regions that are predicted to be intracellular based on the epitope tag data (amino acids 1-352, 441-501, and 868-933), and as predicted, the mAb only detected the protein in permeabilized cells. NGEP is a glycoprotein with predicted glycosylation sites at N809 and N824. When these residues were converted to glutamine, glycosylation was abolished, confirming that the residues are extracellular. Our findings on the expression and the orientation of the NGEP protein serve as an important framework for the development of mAb targeting the extracellular regions of NGEP that could be used for prostate cancer immunotherapy.

    View details for DOI 10.1158/0008-5472.CAN-08-0870

    View details for Web of Science ID 000258177600035

    View details for PubMedID 18676855

    View details for PubMedCentralID PMC2562772

  • The current state of preclinical prostate cancer animal models PROSTATE Pienta, K. J., Abate-Shen, C., Agus, D. B., Attar, R. M., Chung, L. W., Greenberg, N. M., Hahn, W. C., Isaacs, J. T., Navone, N. M., Peehl, D. M., Simons, J. W., Solit, D. B., Soule, H. R., VanDyke, T. A., Weber, M. J., Wu, L., Vessella, R. L. 2008; 68 (6): 629-639

    Abstract

    Prostate cancer continues to be a major cause of morbidity and mortality in men around the world. The field of prostate cancer research continues to be hindered by the lack of relevant preclinical models to study tumorigenesis and to further development of effective prevention and therapeutic strategies. The Prostate Cancer Foundation held a Prostate Cancer Models Working Group (PCMWG) Summit on August 6th and 7th, 2007 to address these issues. The PCMWG reviewed the state of prostate cancer preclinical models and identified the current limitations of cell line, xenograft and genetically engineered mouse models that have hampered the transition of scientific findings from these models to human clinical trials. In addition the PCMWG identified administrative issues that inhibit the exchange of models and impede greater interactions between academic centers and these centers with industry. The PCMWG identified potential solutions for discovery bottlenecks that include: (1) insufficient number of models with insufficient molecular and biologic diversity to reflect human cancer, (2) a lack of understanding of the molecular events that define tumorigenesis, (3) a lack of tools for studying tumor-host interactions, (4) difficulty in accessing model systems across institutions, and (5) addressing why preclinical studies appear not to be predictive of human clinical trials. It should be possible to apply the knowledge gained molecular and epigenetic studies to develop new cell lines and models that mimic progressive and fatal prostate cancer and ultimately improve interventions.

    View details for DOI 10.1002/pros.20726

    View details for Web of Science ID 000255314800006

    View details for PubMedID 18213636

    View details for PubMedCentralID PMC3681409

  • Calcitriol as a chemopreventive and therapeutic agent in prostate cancer: Role of anti-inflammatory activity Conference on the Contemporary Diagnosis and Treatment of Vitamin D-Related Disorders Krishnan, A. V., Moreno, J., Nonn, L., Swami, S., Peehl, D. M., Feldman, D. AMER SOC BONE & MINERAL RES. 2007: V74–V80

    Abstract

    Calcitriol, the hormonally active form of vitamin D, inhibits the growth and development of several cancers. Inflammation has been implicated in the development and progression of many cancers, including prostate cancer (PCa). Recent research from our laboratory suggests that calcitriol exhibits anti-inflammatory actions that may contribute to its inhibitory effects in PCa. We found that calcitriol inhibits the synthesis and actions of pro-inflammatory prostaglandins (PGs) by three mechanisms: (1) inhibition of the expression of cyclooxygenase-2 (COX-2), the enzyme that synthesizes PGs, (2) induction of the expression of 15-prostaglandin dehydrogenase (15-PGDH), the enzyme that inactivates PGs, and (3) decreasing the expression of prostaglandin E and prostaglandin F PG receptors, which are the mediators of PG signaling. The combination of calcitriol and nonsteroidal anti-inflammatory drugs (NSAIDs) result in a synergistic inhibition of PCa cell growth and offers a potential therapeutic strategy. Acting on a separate anti-inflammatory pathway, calcitriol induces the expression of mitogen-activated protein kinase phosphatase 5 (MKP5), a member of a family of phosphatases that are negative regulators of MAP kinases, causing the selective dephosphorylation and inactivation of the stress-activated protein kinase p38. Because p38 activation may be both procarcinogenic and promote inflammation, this calcitriol action, especially coupled with the inhibition of the PG pathway, may contribute to the chemopreventive activity of calcitriol. We conclude that calcitriol exerts several anti-inflammatory actions in prostate cells, which contribute to its potential as a chemopreventive and therapeutic agent in PCa.

    View details for Web of Science ID 000253417900016

    View details for PubMedID 18290727

  • Vitamin D inhibition of the prostaglandin pathway as therapy for prostate cancer Conference on Vitamin D and Cancer - Current Dilemas/Future Needs Feldman, D., Krishnan, A., Moreno, J., Swami, S., Peehl, D. M., Srinivas, S. BLACKWELL PUBLISHING. 2007: S113–S115

    View details for PubMedID 17867384

  • Potentiation of the growth-inhibitory effects of vitamin D in prostate cancer by genistein Conference on Vitamin D and Cancer - Current Dilemas/Future Needs Krishnan, A. V., Swami, S., Moreno, J., Bhattacharyya, R. B., Peehl, D. M., Feldman, D. BLACKWELL PUBLISHING. 2007: S121–S123

    View details for Web of Science ID 000248995500015

    View details for PubMedID 17867387

  • Chemopreventive anti-inflammatory activities of curcumin and other phytochemicals mediated by MAP kinase phosphatase-5 in prostate cells CARCINOGENESIS Nonn, L., Duong, D., Peehl, D. M. 2007; 28 (6): 1188-1196

    Abstract

    As inflammation emerges as a risk factor for prostate cancer (PCa), there is potential for chemoprevention by anti-inflammatory agents. Dietary phytochemicals have been shown to have chemopreventive properties which may include anti-inflammatory activities. In this study, we demonstrate a role for mitogen-activated protein kinase phosphatase-5 (MKP5) in mediating anti-inflammatory activities of the phytochemicals curcumin, resveratrol and [6]-gingerol. We utilized the cytokines tumor necrosis factor-alpha (TNFalpha) and interleukin (IL)-1beta to increase p38-dependent nuclear factor kappa-B (NFkappaB) activation and expression of pro-inflammatory genes cyclooxygenase-2 (COX-2), IL-6 and IL-8 in normal prostatic epithelial cells. MKP5 over-expression decreased cytokine-induced NFkappaB activation, COX-2, IL-6 and IL-8 in normal prostatic epithelial cells, suggesting potent anti-inflammatory activity of MKP5. Pretreatment of cells with a p38 inhibitor mimicked the results observed with MKP5 over-expression, further implicating p38 inhibition as the main activity of MKP5. Curcumin, the phytochemical found in turmeric, up-regulated MKP5, subsequently decreasing cytokine-induced p38-dependent pro-inflammatory changes in normal prostatic epithelial cells. Resveratrol and [6]-gingerol, phytochemicals present in red wine and ginger, respectively, also up-regulated MKP5 in normal prostate epithelial cells. Moreover, we found that PCa cell lines DU 145, PC-3, LNCaP and LAPC-4 retained the ability to up-regulate MKP5 following curcumin, resveratrol and [6]-gingerol exposure, suggesting utility of these phytochemicals in PCa treatment. In summary, our findings show direct anti-inflammatory activity of MKP5 in prostate cells and suggest that up-regulation of MKP5 by phytochemicals may contribute to their chemopreventive actions by decreasing prostatic inflammation.

    View details for DOI 10.1093/carcin/bgl241

    View details for Web of Science ID 000247999700008

    View details for PubMedID 17151092

  • Human prostate epithelium lacks Wee1A-mediated DNA damage-induced checkpoint enforcement PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Hallstrom, T. M., Jaamaa, S., Monkkonen, M., Peltonen, K., Andersson, L. C., Medema, R. H., Peehl, D. M., Laiho, M. 2007; 104 (17): 7211-7216

    Abstract

    Cellular DNA damage triggers the DNA damage response pathway and leads to enforcement of cell cycle checkpoints, which are essential for the maintenance of genomic integrity and are activated in early stages of tumorigenesis. A special feature of prostate cancer is its high incidence and multifocality. To address the functionality of DNA damage checkpoints in the prostate, we analyzed the responses of human primary prostate epithelial cells (HPECs) and freshly isolated human prostate tissues to gamma-irradiation. We find that gamma-irradiation activates the ataxia telangiectasia mutated-associated DNA damage response pathway in the HPECs but that the clearance of phosphorylated histone H2AX (gammaH2AX) foci is delayed. Surprisingly, gamma-irradiated HPECs were unable to enforce cell cycle checkpoint arrest and had sustained cyclin-dependent kinase 2 (Cdk2)-associated kinase activity because of a lack of inhibitory Cdk phosphorylation by Wee1A tyrosine kinase. We further show that HPECs express low levels of Wee1A and that ectopic Wee1A efficiently rescues the checkpoints. We recapitulate the absence of checkpoint responses in epithelium of ex vivo irradiated human prostate tissue despite robust induction of gammaH2AX. The findings show that prostate epithelium has a surprising inability to control checkpoint arrest, the lack of which may predispose to accrual of DNA lesions.

    View details for DOI 10.1073/pnas.0609299104

    View details for Web of Science ID 000246024700059

    View details for PubMedID 17431037

    View details for PubMedCentralID PMC1855358

  • Novel pathways that contribute to the anti-proliferative and chemopreventive activities of calcitriol in prostate cancer 13th Workshop on Vitamin D Krishnan, A. V., Moreno, J., Nonn, L., Malloy, P., Swami, S., Peng, L., Peehl, D. M., Feldman, D. PERGAMON-ELSEVIER SCIENCE LTD. 2007: 694–702

    Abstract

    Calcitriol, the hormonally active form of Vitamin D, inhibits the growth and development of many cancers through multiple mechanisms. Our recent research supports the contributory role of several new and diverse pathways that add to the mechanisms already established as playing a role in the actions of calcitriol to inhibit the development and progression of prostate cancer (PCa). Calcitriol increases the expression of insulin-like growth factor binding protein-3 (IGFBP-3), which plays a critical role in the inhibition of PCa cell growth by increasing the expression of the cell cycle inhibitor p21. Calcitriol inhibits the prostaglandin (PG) pathway by three actions: (i) the inhibition of the expression of cyclooxygenase-2 (COX-2), the enzyme that synthesizes PGs, (ii) the induction of the expression of 15-prostaglandin dehydrogenase (15-PGDH), the enzyme that inactivates PGs and (iii) decreasing the expression of EP and FP PG receptors that are essential for PG signaling. Since PGs have been shown to promote carcinogenesis and progression of multiple cancers, the inhibition of the PG pathway may add to the ability of calcitriol to prevent and inhibit PCa development and growth. The combination of calcitriol and non-steroidal anti-inflammatory drugs (NSAIDs) result in a synergistic inhibition of PCa cell growth and offers a potential therapeutic strategy. Mitogen activated protein kinase phosphatase 5 (MKP5) is a member of a family of phosphatases that are negative regulators of MAP kinases. Calcitriol induces MKP5 expression in prostate cells leading to the selective dephosphorylation and inactivation of the stress-activated kinase p38. Since p38 activation is pro-carcinogenic and is a mediator of inflammation, this calcitriol action, especially coupled with the inhibition of the PG pathway, contributes to the chemopreventive activity of calcitriol in PCa. Mullerian Inhibiting Substance (MIS) has been evaluated for its inhibitory effects in cancers of the reproductive tissues and is in development as an anti-cancer drug. Calcitriol induces MIS expression in prostate cells revealing yet another mechanism contributing to the anti-cancer activity of calcitriol in PCa. Thus, we conclude that calcitriol regulates myriad pathways that contribute to the potential chemopreventive and therapeutic utility of calcitriol in PCa.

    View details for DOI 10.1016/j.jsbmb.2006.12.051

    View details for Web of Science ID 000245826800094

    View details for PubMedID 17229571

  • Calcitriol and genistein actions to inhibit the prostaglandin pathway: Potential combination therapy to treat prostate cancer International Research Conference on Food, Nutrition, and Cancer Swami, S., Krishnan, A. V., Moreno, J., Bhattacharyya, R. B., Peehl, D. M., Feldman, D. AMER SOC NUTRITIONAL SCIENCE. 2007: 205S–210S

    Abstract

    We present an overview of the prostaglandin (PG) pathway as a novel target for the treatment of prostate cancer (PCa) using a combination of calcitriol and genistein, both of which have known antiproliferative properties. Calcitriol inhibits the PG pathway in PCa cells in 3 separate ways: by decreasing cyclooxygenase-2 (COX-2) expression, stimulating 15-hydroxyprostaglandin dehydrogenase (15-PGDH) expression, and decreasing EP (PGE2) and FP (PGF(2alpha)) receptors. These actions of calcitriol result in reduced levels of biologically active PGE2, leading ultimately to growth inhibition of the PCa cells. We also demonstrate the advantages of using calcitriol in combination with genistein for the treatment of PCa. Genistein, a major component of soy, is a potent inhibitor of the activity of CYP24, the enzyme that initiates the degradation of calcitriol. This leads to increased half-life of bioactive calcitriol, thereby enhancing all of calcitriol's actions including those on the PG pathway. In addition to inhibiting CYP24 enzyme activity, genistein has its own independent actions on the PG pathway in PCa cells. Like calcitriol it inhibits COX-2 expression and activity, leading to decreased synthesis of PGE2. It also inhibits the EP and FP receptors, thereby reducing the biological function of PGE2. Thus, the combination of calcitriol and genistein acts additively to inhibit the PG pathway. Both calcitriol and genistein are relatively safe and have little toxicity associated with their intake. We postulate that the combination of calcitriol and genistein is an attractive therapeutic option for the treatment of PCa.

    View details for Web of Science ID 000243330800009

    View details for PubMedID 17182827

  • High resolution regional elasticity mapping of the human prostate 29th Annual International Conference of the IEEE-Engineering-in-Medicine-and-Biology-Society Murayama, Y., Omata, S., Yajima, T., Peng, Q., Shishido, K., Peehl, D. M., Constantinou, C. E. IEEE. 2007: 5803–5806

    Abstract

    What is it that the clinician "feels" during a digital rectal examination? To answer this question, it is necessary to measure the elastic properties of the prostate and verify the stiffness values with histological examination. Therefore, we devised an Elasticity Mapping System to evaluate the elastic properties of various histopathological grades of prostate cancer in relation to benign prostatic hyperplasia (BPH) and normal tissue. The system consists of a micro tactile sensor, a three-axis (XYZ) with one (fine Z) micromanipulation stage, a stereoscope camera and a measurement chamber. Using this methodology we mapped the elasticity of human prostate cancer (CaP) and it was obviously observed that the node was significantly harder than surrounding normal tissues and had some textures.

    View details for Web of Science ID 000253467004188

    View details for PubMedID 18003332

  • Distinctive gene expression of prostatic stromal cells cultured from diseased versus normal tissues JOURNAL OF CELLULAR PHYSIOLOGY Zhao, H., Ramos, C. F., Brooks, J. D., Peehl, D. M. 2007; 210 (1): 111-121

    Abstract

    To obtain a comprehensive view of the transcriptional programs in prostatic stromal cells of different histological/pathological origin, we profiled 18 adult human stromal cell cultures from normal transition zone (TZ), normal peripheral zone (PZ), benign prostatic hyperplasia (BPH), and prostate cancer (CA) using cDNA microarrays. A hierarchical clustering analysis of 714 named unique genes whose expression varied at least threefold from the overall mean abundance in at least three samples in all 18 samples demonstrated that cells of different origin displayed distinct gene expression profiles. Many of the differentially expressed genes are involved in biological processes known to be important in the development of prostatic diseases including cell proliferation and apoptosis, cell adhesion, and immune response. Significance Analysis of Microarrays (SAM) analysis identified genes that showed differential expression with statistical significance including 24 genes between cells from TZ versus BPH, 34 between BPH versus CA, and 101 between PZ versus CA. S100A4 and SULF1, the most up- and downregulated genes in BPH versus TZ, respectively, showed expression at the protein level consistent with microarray analysis. In addition, sulfatase assay showed that BPH cells have lower SULF1 activity compared to TZ cells. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis confirmed differential expression of ENPP2/autotoxin and six other genes between PZ versus CA, as well as differential expression of six genes between BPH versus CA. Our results support the hypothesis that prostatic stromal cells of different origin have unique transcriptional programs and point towards genes involved in actions of stromal cells in BPH and CA.

    View details for DOI 10.1002/jcp.20828

    View details for Web of Science ID 000242568200012

    View details for PubMedID 17044071

    View details for PubMedCentralID PMC2732006

  • Transcript profiling of the androgen signal in normal prostate, benign prostatic hyperplasia, and prostate cancer ENDOCRINOLOGY Bauman, D. R., Steckelbroeck, S., Peehl, D. M., Penning, T. M. 2006; 147 (12): 5806-5816

    Abstract

    Human prostate adenocarcinoma (CaP) and benign prostatic hyperplasia (BPH) have epithelial and stromal cell origins, respectively. To determine whether the androgen signal is processed differently in these cell types the expression of transcripts for enzymes that control ligand access to the androgen receptor (AR) were measured. Transcripts for type 2 5alpha-reductase, ketosteroid reductases [aldo-keto reductase (AKR)1C1-AKR1C4], the major oxidative 3alpha-hydroxysteroid dehydrogenase (HSD) retinol dehydrogenase (RODH)-like 3alpha-HSD (RL-HSD) and nuclear receptors [AR, estrogen receptor (ER)alpha, and ERbeta] were determined in whole human prostate and in cultures of primary epithelial cells (PEC) and primary stromal cells (PSC) from normal prostate, CaP and BPH by real-time RT-PCR. Normal PEC (n=14) had higher levels of AKR1C1 (10-fold, P<0.001), AKR1C2 (115-fold, P<0.001) and AKR1C3 (6-fold, P<0.001) than normal PSC (n=15), suggesting that reductive androgen metabolism occurs. By contrast, normal PSC had higher levels of AR (8-fold, P<0.001) and RL-HSD (21-fold, P<0.001) than normal PEC, suggesting that 3alpha-androstanediol is converted to 5alpha-dihydrotestosterone to activate AR. In CaP PEC (n=14), no significant changes in transcript levels vs. normal PEC were observed. In BPH PSC (n=21) transcripts for AR (2-fold, P<0.001), AKR1C1 (4-fold, P<0.001), AKR1C2 (10-fold P<0.001), AKR1C3 (4-fold, P<0.001) and RL-HSD (3-fold, P<0.003) were elevated to increase androgen response. Differences in the AR:ERbeta transcript ratios (eight in normal PEC vs. 280 in normal PSC) were maintained in PEC and PSC in diseased prostate. These data suggest that CaP may be more responsive to an ERbeta agonist and BPH may be more responsive to androgen ablation.

    View details for DOI 10.1210/en.2006-0627

    View details for Web of Science ID 000242047200034

    View details for PubMedID 16959841

  • Mechanisms of vitamin D-mediated growth inhibition in prostate cancer cells: Inhibition of the prostaglandin pathway 2nd International Symposium on Vitamin D Analogs in Cancer Prevention and Therapy Moreno, J., Krishnan, A. V., Peehl, D. M., Feldman, D. INT INST ANTICANCER RESEARCH. 2006: 2525–30

    Abstract

    Calcitriol (1,25-dihydroxyvitamin D3), the active form of vitamin D, promotes growth inhibition and differentiation in prostate cancer (PCa) cells. To unravel the molecular pathways of calcitriol actions, cDNA microarray analysis was used to identify novel calcitriol target genes including two that play key roles in the metabolism of prostaglandins (PGs), known stimulators of PCa growth and progression. Calcitriol significantly decreases the expression of the PG synthesizing cyclooxygenase-2 (COX-2) gene, while increasing that of PG inactivating 15-prostaglandin dehydrogenase (15-PGDH). Calcitriol also inhibits the expression of the PG receptors EP2 and FP. It reduces the levels of biologically active PGs and inhibits PG actions in PCa cells, thereby decreasing the proliferative stimulus of PGs. We postulate that the regulation of the PG pathway contributes to the growth inhibitory actions of calcitriol. We also propose that calcitriol can be combined with non-steroidal anti-inflammatory drugs (NSAIDs) that inhibit COX enzyme activity, as a potential therapeutic strategy in PCa.

    View details for Web of Science ID 000241391500003

    View details for PubMedID 16886660

  • Inhibition of p38 by vitamin D reduces interleukin-6 production in normal prostate cells via mitogen-activated protein kinase phosphatase 5: Implications for prostate cancer prevention by vitamin D CANCER RESEARCH Nonn, L., Peng, L. H., Feldman, D., Peehl, D. M. 2006; 66 (8): 4516-4524

    Abstract

    Although numerous studies have implicated vitamin D in preventing prostate cancer, the underlying mechanism(s) remains unclear. Using normal human prostatic epithelial cells, we examined the role of mitogen-activated protein kinase phosphatase 5 (MKP5) in mediating cancer preventive activities of vitamin D. Up-regulation of MKP5 mRNA by 1,25-dihydroxyvitamin-D3 (1,25D) was dependent on the vitamin D receptor. We also identified a putative positive vitamin D response element within the MKP5 promoter that associated with the vitamin D receptor following 1,25D treatment. MKP5 dephosphorylates/inactivates the stress-activated protein kinase p38. Treatment of prostate cells with 1,25D inhibited p38 phosphorylation, and MKP5 small interfering RNA blocked this effect. Activation of p38 and downstream production of interleukin 6 (IL-6) are proinflammatory. Inflammation and IL-6 overexpression have been implicated in the initiation and progression of prostate cancer. 1,25D pretreatment inhibited both UV- and tumor necrosis factor alpha-stimulated IL-6 production in normal cells via p38 inhibition. Consistent with inhibition of p38, 1,25D decreased UV-stimulated IL-6 mRNA stabilization. The ability of 1,25D to up-regulate MKP5 was maintained in primary prostatic adenocarcinoma cells but was absent in metastases-derived prostate cancer cell lines. The inability of 1,25D to regulate MKP5 in the metastasis-derived cancer cells suggests there may be selective pressure to eliminate key tumor suppressor functions of vitamin D during cancer progression. These studies reveal MKP5 as a mediator of p38 inactivation and decreased IL-6 expression by 1,25D in primary prostatic cultures of normal and adenocarcinoma cells, implicating decreased prostatic inflammation as a potential mechanism for prostate cancer prevention by 1,25D.

    View details for DOI 10.1158/0008-5472.CAN-05-3796

    View details for Web of Science ID 000236843200071

    View details for PubMedID 16618780

  • Vitamin D selectively decreases DNA damage in normal human primary prostate cells but not in prostate cancer cell lines Nonn, L., Peehl, D. M. AMER ASSOC CANCER RESEARCH. 2006
  • Aldo-keto reductase (AKR) 1C3: Role in prostate disease and the development of specific inhibitors International Workshop on 11beta and 17beta-Hydroxysteroid Dehydrogenases Penning, T. M., Steckelbroeck, S., Bauman, D. R., Miller, M. W., Jin, Y., Peehl, D. M., Fung, K. M., Lin, H. K. ELSEVIER IRELAND LTD. 2006: 182–91

    Abstract

    Human aldo-keto reductases (AKR) of the 1A, 1B, 1C and 1D subfamilies are involved in the pre-receptor regulation of nuclear (steroid hormone and orphan) receptors by regulating the local concentrations of their lipophilic ligands. AKR1C3 is one of the most interesting isoforms. It was cloned from human prostate and the recombinant protein was found to function as a 3-, 17- and 20-ketosteroid reductase with a preference for the conversion of Delta4-androstene-3,17-dione to testosterone implicating this enzyme in the local production of active androgens within the prostate. Using a validated isoform specific real-time RT-PCR procedure the AKR1C3 transcript was shown to be more abundant in primary cultures of epithelial cells than stromal cells, and its expression in stromal cells increased with benign and malignant disease. Using a validated isoform specific monoclonal Ab, AKR1C3 protein expression was also detected in prostate epithelial cells by immunoblot analysis. Immunohistochemical staining of prostate tissue showed that AKR1C3 was expressed in adenocarcinoma and surprisingly high expression was observed in the endothelial cells. These cells are a rich source of prostaglandin G/H synthase 2 (COX-2) and vasoactive prostaglandins (PG) and thus the ability of recombinant AKR1C enzymes to act as PGF synthases was compared. AKR1C3 had the highest catalytic efficiency (kcat/Km) for the 11-ketoreduction of PGD2 to yield 9alpha,11beta-PGF2 raising the prospect that AKR1C3 may govern ligand access to peroxisome proliferator activated receptor (PPARgamma). Activation of PPARgamma is often a pro-apoptotic signal and/or leads to terminal differentiation, while 9alpha,11beta-PGF2 is a pro-proliferative signal. AKR1C3 is potently inhibited by non-steroidal anti-inflammatory drugs suggesting that the cancer chemopreventive properties of these agents may be mediated either by inhibition of AKR1C3 or COX. To discriminate between these effects we developed potent AKR1C inhibitors based on N-phenylanthranilic acids that do not inhibit COX-1 or COX-2. These compounds can now be used to determine the role of AKR1C3 in producing two proliferative signals in the prostate namely testosterone and 9alpha,11beta-PGF2.

    View details for DOI 10.1016/j.mce.2005.12.009

    View details for Web of Science ID 000236691100028

    View details for PubMedID 16417966

  • Tumor-selective killing by selenite in patient-matched pairs of normal and malignant prostate cells PROSTATE Husbeck, B., Nonn, L., Peehl, D. M., Knox, S. J. 2006; 66 (2): 218-225

    Abstract

    Selenium compounds have been shown to induce apoptosis in a variety of human prostate cancer cell lines. However, the effects of selenium have yet to be examined in normal and malignant cells derived from the same individual. Selenite metabolism consumes glutathione (GSH) and produces superoxide. The generation of reactive oxygen species is an important mechanism in selenite-induced apoptosis.Three patient-matched pairs of primary prostatic epithelial cell cultures from normal and cancer were evaluated for their response to selenite. Apoptosis was measured and the differential response of normal and cancer cells was correlated with the expression of bcl-2, bax, GSH, and manganese superoxide dismutase (MnSOD).The cancer-derived cells were significantly more sensitive to selenite-induced apoptosis than the corresponding normal cells. Tumor-selective killing was not observed in cells treated with selenomethionine. The ratio of bcl-2:bax was decreased in the cancer-derived cells treated with selenite. Total GSH concentrations were similar in paired normal and cancer cells. Therefore, differences in GSH content do not appear to play a role in tumor-selective killing by selenite. Superoxide is a by-product of selenite metabolism and normal cells showed increased MnSOD expression and SOD activity compared to the cancer-derived cells. Prostate cancer cells treated with the MnSOD mimetic, MnTMPyP, were protected against the cytotoxic effects of selenite.Higher MnSOD expression in normal cells may play an important role in eliminating superoxide radicals produced as a result of selenite metabolism and contribute to the tumor-selective killing by selenite in prostate cancer.

    View details for DOI 10.1002/pros.20337

    View details for Web of Science ID 000234838300011

    View details for PubMedID 16173037

  • Expression of CCL5 (RANTES) and CCR5 in prostate cancer PROSTATE Vaday, G. G., Peehl, D. M., Kadam, P. A., Lawrence, D. M. 2006; 66 (2): 124–34

    Abstract

    Expression of the inflammatory chemokine CCL5 (RANTES) by tumor cells is thought to correlate with the progression of several cancers. CCL5 was shown to induce breast cancer cell migration, mediated by the receptor CCR5. A CCR5 antagonist was demonstrated to inhibit experimental breast tumor growth. Recently, CCL5 and CCR5 mRNA expression was reported in prostate cancer (PCa) tissues. Herein, we characterized CCL5 and CCR5 expression in cultures of PCa cells and explored possible functions of CCL5 in PCa progression.Quantitative RT-PCR, ELISA, and immunohistochemical staining were performed to examine CCL5 expression in prostate cell lines. CCR5 expression was measured by flow cytometry. Proliferation and invasion assays were performed to determine potential functions of CCL5 and CCR5 in PCa.Expression of CCL5 mRNA and protein was found in human PCa cell lines (PC-3; DU-145; LNCaP) and primary prostate adenocarcinoma cells. CCL5 and CCR5 were also detected in human PCa tissues. CCR5 expression was demonstrated on the cell surface of PCa cells, as well as in intracellular pools. Incubation with CCL5 (10-100 ng/ml) induced PCa cell proliferation, and the CCR5 antagonist TAK-779 inhibited CCL5-induced proliferation. CCL5 was found to stimulate PCa cell invasion, and TAK-779 blocked the effects of CCL5.In light of evidence that inflammation influences the pathogenesis of PCa, these results suggest that inflammatory chemokines, such as CCL5, expressed by prostate cells may act directly on the growth and survival of PCa cells. Chemokine receptor antagonists may thus block autocrine mechanisms of PCa progression.

    View details for DOI 10.1002/pros.20306

    View details for Web of Science ID 000234838300002

    View details for PubMedID 16161154

  • Identification of the major oxidative 3 alpha-hydroxysteroid dehydrogenase in human prostate that converts 5 alpha-androstane-3 alpha,17 beta-diol to 5 alpha-dihydrotestosterone: A potential therapeutic target for androgen-dependent disease MOLECULAR ENDOCRINOLOGY Bauman, D. R., Steckelbroeck, S., Williams, M. V., Peehl, D. M., Penning, T. M. 2006; 20 (2): 444-458

    Abstract

    Androgen-dependent prostate diseases initially require 5alpha-dihydrotestosterone (DHT) for growth. The DHT product 5alpha-androstane-3alpha,17beta-diol (3alpha-diol), is inactive at the androgen receptor (AR), but induces prostate growth, suggesting that an oxidative 3alpha-hydroxysteroid dehydrogenase (HSD) exists. Candidate enzymes that posses 3alpha-HSD activity are type 3 3alpha-HSD (AKR1C2), 11-cis retinol dehydrogenase (RODH 5), L-3-hydroxyacyl coenzyme A dehydrogenase , RODH like 3alpha-HSD (RL-HSD), novel type of human microsomal 3alpha-HSD, and retinol dehydrogenase 4 (RODH 4). In mammalian transfection studies all enzymes except AKR1C2 oxidized 3alpha-diol back to DHT where RODH 5, RODH 4, and RL-HSD were the most efficient. AKR1C2 catalyzed the reduction of DHT to 3alpha-diol, suggesting that its role is to eliminate DHT. Steady-state kinetic parameters indicated that RODH 4 and RL-HSD were high-affinity, low-capacity enzymes whereas RODH 5 was a low-affinity, high-capacity enzyme. AR-dependent reporter gene assays showed that RL-HSD, RODH 5, and RODH 4 shifted the dose-response curve for 3alpha-diol a 100-fold, yielding EC(50) values of 2.5 x 10(-9) M, 1.5 x 10(-9) M, and 1.0 x 10(-9) M, respectively, when compared with the empty vector (EC(50) = 1.9 x 10(-7) M). Real-time RT-PCR indicated that L-3-hydroxyacyl coenzyme A dehydrogenase and RL-HSD were expressed more than 15-fold higher compared with the other candidate oxidative enzymes in human prostate and that RL-HSD and AR were colocalized in primary prostate stromal cells. The data show that the major oxidative 3alpha-HSD in normal human prostate is RL-HSD and may be a new therapeutic target for treating prostate diseases.

    View details for DOI 10.1210/me.2005-0287

    View details for Web of Science ID 000235129600018

    View details for PubMedID 16179381

  • Tumor-suppression function of transcription factor USF2 in prostate carcinogenesis ONCOGENE Chen, N., Szentirmay, M. N., Pawar, S. A., Sirito, M., Wang, J., Wang, Z., Zhai, Q., Yang, H. X., Peehl, D. M., Ware, J. L., Sawadogo, M. 2006; 25 (4): 579-587

    Abstract

    Although the transcription factor USF2 has been implicated in the regulation of cellular growth and proliferation, it is unknown whether alterations in USF2 contribute to tumorigenesis and tumor development. We examined the role of USF2 in prostate tumorigenesis. Western blot analysis revealed markedly decreased USF2 levels in three androgen-independent prostate cancer cell lines, PC-3, DU145, and M12, as compared to nontumorigenic prostate epithelial cells or the androgen-dependent cell line, LNCaP. Ectopic expression of USF2 in PC-3 cells did not affect the cell proliferation rate of PC-3 cells on plastic surfaces. However, it dramatically decreased anchorage-independent growth of PC-3 cells in soft agar (90-98% inhibition) and the invasion capability (80% inhibition) of PC-3 cells in matrix gel assay. Importantly, expression of USF2 in PC-3 cells inhibited the tumorigenicity of PC-3 cells in an in vivo nude mice xenograft model (80-90% inhibition). These results suggest that USF2 has tumor-suppression function. Consistent with its function in tumor suppression, we found that the USF2 protein is present in normal prostate epithelial cells but absent in 18 of 42 (43%) human prostate cancer tissues (P = 0.015). To further examine the functional role of USF2 in vivo, we generated mice with genetic deletion of USF2 gene. We found that USF2-null mice displayed marked prostate hyperplasia at a young age, suggesting that USF2 is involved in the normal growth and differentiation of prostate. Together, these studies demonstrate that USF2 has tumor-suppressor function and plays a role in prostate carcinogenesis.

    View details for Web of Science ID 000234897400009

    View details for PubMedID 16186802

  • Genistein potentiates the growth inhibitory effects of 1,25-dihydroxyvitamin D-3 in DU145 human prostate cancer cells: Role of the direct inhibition of CYP24 enzyme activity MOLECULAR AND CELLULAR ENDOCRINOLOGY Swami, S., Krishnan, A. V., Peehl, D. M., Feldman, D. 2005; 241 (1-2): 49-61

    Abstract

    In a search for improved therapies for prostate cancer, we investigated the effect of genistein in combination with 1alpha-25-dihydroxyvitamin D3 [1,25(OH)2D3], on the growth of DU145 human prostate cancer cells. DU145 cells were very resistant to the growth inhibitory action of 1,25(OH)2D3 or genistein when administered individually. However, the combination caused a significant growth inhibition seen at lower concentrations of both agents. 1,25(OH)2D3 induces the expression of the CYP24 gene, which codes for the enzyme that initiates the catabolism of 1,25(OH)2D3. We showed for the first time that genistein at low doses (50-100 nM) directly inhibited CYP24 at the enzyme level. Addition of genistein to mitochondrial preparations inhibited CYP24 enzyme activity in a noncompetitive manner. CYP24 inhibition by genistein increased the half-life of 1,25(OH)2D3 thereby augmenting the homologous up-regulation of the vitamin D receptor (VDR) both at the mRNA and protein levels. Genistein co-treatment enhanced 1,25(OH)2D3-mediated transactivation of the vitamin D responsive reporters OC-Luc and OP-Luc transfected into DU145 cells. Consistent with the growth inhibition due to the combination treatment, significant changes in the expression of genes involved in growth arrest and apoptosis were seen. We conclude that genistein potentiates the antiproliferative actions of 1,25(OH)2D3 in DU145 cells by two mechanisms: (i) an increase in the half-life of 1,25(OH)2D3 due to the direct inhibition of CYP24 enzyme activity and (ii) an amplification of the homologous up-regulation of VDR. Together these two effects lead to a substantial enhancement of the cellular responses to the growth inhibitory and pro-apoptotic signaling by 1,25(OH)2D3.

    View details for DOI 10.1016/j.mce.2005.05.001

    View details for Web of Science ID 000232439700006

    View details for PubMedID 15955619

  • Comparative gene and protein expression in primary cultures of epithelial cells from benign prostatic hyperplasia and prostate cancer CANCER LETTERS Rose, A., Xu, Y., Chen, Z. X., Fan, Z. B., Stamey, T. A., McNeal, J. E., Caldwell, M., Peehl, D. M. 2005; 227 (2): 213-222

    Abstract

    Primary cultures are widely used to investigate the disease-specific biology of prostate cancer and benign prostatic hyperplasia (BPH). To identify genes differentially expressed between epithelial cells cultured from adenocarcinomas versus BPH tissues, we used probe array technology. Gene expression profiles were evaluated on Affymetrix Human Cancer G110 Array Chips containing approximately 1900 cancer-related genes. After defined statistical analysis, genes that were over-expressed in cancer cultures were identified. Protein expression of four of the differentially expressed genes was measured in immunoblots, and the expression of two other genes was measured by real-time reverse transcription-polymerase chain reaction (RT-PCR). While no gene or protein was consistently over-expressed in all cancer versus BPH cell cultures, cytokeratin 16 protein was highly elevated in several of the cancer cultures, suggesting that a hyperproliferative phenotype may be characteristic of prostate cancer cells.

    View details for DOI 10.1016/j.canlet.2005.01.037

    View details for PubMedID 16112424

  • Regulation of prostaglandin metabolism by calcitriol attenuates growth stimulation in prostate cancer cells CANCER RESEARCH Moreno, J., Krishnan, A. V., Swami, S., Nonn, L., Peehl, D. M., Feldman, D. 2005; 65 (17): 7917-7925

    Abstract

    Calcitriol exhibits antiproliferative and pro-differentiation effects in prostate cancer. Our goal is to further define the mechanisms underlying these actions. We studied established human prostate cancer cell lines and primary prostatic epithelial cells and showed that calcitriol regulated the expression of genes involved in the metabolism of prostaglandins (PGs), known stimulators of prostate cell growth. Calcitriol significantly repressed the mRNA and protein expression of prostaglandin endoperoxide synthase/cyclooxygenase-2 (COX-2), the key PG synthesis enzyme. Calcitriol also up-regulated the expression of 15-hydroxyprostaglandin dehydrogenase, the enzyme initiating PG catabolism. This dual action was associated with decreased prostaglandin E2 secretion into the conditioned media of prostate cancer cells exposed to calcitriol. Calcitriol also repressed the mRNA expression of the PG receptors EP2 and FP, providing a potential additional mechanism of suppression of the biological activity of PGs. Calcitriol treatment attenuated PG-mediated functional responses, including the stimulation of prostate cancer cell growth. The combination of calcitriol with nonsteroidal anti-inflammatory drugs (NSAIDs) synergistically acted to achieve significant prostate cancer cell growth inhibition at approximately 2 to 10 times lower concentrations of the drugs than when used alone. In conclusion, the regulation of PG metabolism and biological actions constitutes a novel pathway of calcitriol action that may contribute to its antiproliferative effects in prostate cells. We propose that a combination of calcitriol and nonselective NSAIDs might be a useful chemopreventive and/or therapeutic strategy in men with prostate cancer, as it would allow the use of lower concentrations of both drugs, thereby reducing their toxic side effects.

    View details for DOI 10.1158/0008-5472.CAN-05-1435

    View details for Web of Science ID 000231659500052

    View details for PubMedID 16140963

  • Molecular targets of Doxazosin in human prostatic stromal cells PROSTATE Zhao, H. J., Lai, F., Nonn, L., Brooks, J. D., Peehl, D. M. 2005; 62 (4): 400-410

    Abstract

    We used cDNA microarray analysis to obtain insights into the mechanisms of action of doxazosin, an alpha(1)-adrenergic receptor antagonist used to treat benign prostatic hyperplasia (BPH).Hierarchical clustering analysis and significance analysis of microarray (SAM) were performed to identify genes differentially expressed between untreated stromal cells cultured from normal tissue and BPH, and changes in gene expression induced by doxazosin. Transcript levels of selected genes were validated by real-time reverse-transcription polymerase chain reaction (RT-PCR).Hierarchical clustering analyses separated untreated normal and BPH cells. Sixty-seven genes whose expression varied at least twofold after doxazosin treatment in both normal and BPH cells were identified, as were 93 genes differentially regulated in normal versus BPH cells. Molecular targets consistent with tumor necrosis factor (TNF)-alpha-related activity were identified.Normal versus BPH stromal cells differ in global gene transcription. Doxazosin induced gene expression changes relevant to proliferation/apoptosis, immune defense, cell-cell signaling/signal transduction, and transcriptional regulation.

    View details for DOI 10.1002/pros.20161

    View details for Web of Science ID 000226991500011

    View details for PubMedID 15378519

  • Primary cell cultures as models of prostate cancer development ENDOCRINE-RELATED CANCER Peehl, D. M. 2005; 12 (1): 19-47

    Abstract

    This review focuses on primary cultures of human prostatic epithelial cells and their applications as models of normal and malignant biological behavior. Current abilities to culture cells from normal tissues, from premalignant dysplastic lesions (prostatic intraepithelial neoplasia), from primary adenocarcinomas, and from metastases are described. Evidence for representation of the interrelated cells of the normal prostatic epithelium--stem cells, basal epithelial cells, secretory epithelial cells, transit amplifying cells and neuroendocrine cells--in primary cultures is presented. Comparisons between normal and cancer-derived primary cultures are made regarding biological activities relevant to carcinogenesis, such as proliferation, apoptosis, differentiation, senescence, adhesion, migration, invasion, steroid hormone metabolism, other metabolic pathways and angiogenesis. Analyses of tumor suppressor activity, differential gene expression and cytogenetics in primary cultures have revealed changes relevant to prostate cancer progression. Preclinical studies with primary cultures have provided information useful for designing new strategies for chemoprevention, chemotherapy, cytotoxin therapy, radiation therapy, gene therapy and imaging. While the behavior of normal primary cultures is often used as a basis for comparison with established, immortal prostate cancer cell lines, the most informative studies are performed with donor-matched pairs of normal and malignant primary cultures, grown under identical conditions. Challenges that remain to be addressed if the full potential of primary cultures as a model system is to be realized include isolation, culture and characterization of stem cells, improved methodology to induce or maintain a fully differentiated, androgen-responsive phenotype, and identification of cell surface antigens or other markers with which to purify pure populations of live cancer or premalignant cells apart from non-malignant epithelial cells prior to culture.

    View details for DOI 10.1677/erc.1.00795

    View details for Web of Science ID 000228049800002

    View details for PubMedID 15788637

  • Preferential production of latent transforming growth factor beta-2 by primary prostatic epithelial cells and its activation by prostate-specific antigen JOURNAL OF CELLULAR PHYSIOLOGY Dallas, S. L., Zhao, S., Cramer, S. D., Chen, Z., Peehl, D. M., Bonewald, L. F. 2005; 202 (2): 361-370

    Abstract

    Three mammalian isoforms of transforming growth factor-beta (TGFbeta) are known, TGFbeta1, 2, and 3, that have non-overlapping functions during development. However, their specific roles in cancers such as prostate cancer are less clear. Here we show that primary cultures of prostatic epithelial cells preferentially produce and activate the latent TGFbeta2 isoform. Paired cultures of normal and malignant prostate cells from prostate cancer patients produced predominantly the TGFbeta2 isoform, with 30- to 70-fold less TGFbeta1. By mono-Q ion exchange chromatography, three major peaks of latent TGFbeta2 activity were observed corresponding to the known small latent TGFbeta2 complex, the known large latent TGFbeta2 complex and a novel eluting peak of latent TGFbeta2. Although prostate cells are known to activate latent TGFbeta, the mechanism for activation is currently unclear. We investigated whether prostate specific antigen (PSA), a serine protease used as a clinical marker for prostate cancer, could play a role in the activation of latent TGFbeta. Unlike plasmin, a known activator of both latent TGFbeta1 and 2, PSA specifically activated the recombinant small latent form of TGFbeta2, but not TGFbeta1. Prostate epithelial cells, therefore, preferentially produce the TGFbeta2 isoform and PSA, a protease produced by the prostate, specifically targets the activation of this TGFbeta isoform. PSA-mediated activation of latent TGFbeta2 may be an important mechanism for autocrine TGFbeta regulation in the prostate and may potentially contribute to the formation of osteoblastic lesions in bone metastatic prostate cancer.

    View details for DOI 10.1002/jcp.20147

    View details for Web of Science ID 000226089400005

    View details for PubMedID 15389580

  • Redox modulation of human prostate carcinoma cells by selenite increases radiation-induced cell killing FREE RADICAL BIOLOGY AND MEDICINE Husbeck, B., Peehl, D. M., Knox, S. J. 2005; 38 (1): 50-57

    Abstract

    Although selenium compounds have been extensively studied as chemopreventative agents for prostate cancer, little is known about the potential use of selenium compounds for chemotherapy. We have shown that selenite inhibits cell growth and induces apoptosis in androgen-dependent LAPC-4 prostate cancer cells. LAPC-4 cells were more sensitive to selenite-induced apoptosis than primary cultures of normal prostate cells. Selenite-induced apoptosis in LAPC-4 cells correlated with a decrease in the Bcl-2:Bax expression ratio. Selenite-induced oxidative stress and apoptosis are dependent upon its reaction with reduced GSH. LAPC-4 cells treated with selenite showed decreased levels of total GSH and increased concentrations of GSSG. Thus, selenite altered the intracellular redox status toward an oxidative state by decreasing the ratio of GSH:GSSG. Because increased levels of Bcl-2 and GSH are associated with radioresistance, we examined the ability of selenite to sensitize prostate cancer cells to gamma-irradiation. Both LAPC-4 and androgen-independent DU 145 cells pretreated with selenite showed increased sensitivity to gamma-irradiation as measured by clonogenic survival assays. Importantly, selenite-induced radiosensitization was observed in combination with a clinically relevant dose of 2 Gy. These data suggest that altering the redox environment of prostate cancer cells with selenite increases the apoptotic potential and sensitizes them to radiation-induced cell killing.

    View details for DOI 10.1016/j.freeradbiomed.2004.09.022

    View details for Web of Science ID 000226172900006

    View details for PubMedID 15589371

  • Vitamin D and Prostate Cancer VITAMIN D: VOLS 1 AND 2, 2ND EDITION Krishnan, A. V., Peehl, D. M., Feldman, D., Feldman, D. 2005: 1679–1707
  • A methylation profile of in vitro immortalized human cell lines INTERNATIONAL JOURNAL OF ONCOLOGY Liu, L. M., ZHANG, J. M., BATES, S., Li, J. J., Peehl, D. M., Rhim, J. S., Pfeifer, G. P. 2005; 26 (1): 275-285

    Abstract

    Normal human diploid cells have a limited life span and undergo replicative senescence after various limited population doublings. Cells must pass the senescence barrier to become immortal. The exact mechanisms of immortalization are not clear, although inactivation of the RB pathway, and/or the p53 pathway and activation of telomerase has been shown to be necessary for immortalization of certain cell types with DNA viruses or hTERT. Methylation-associated inactivation of tumor suppressor genes plays an important role in tumor progression. To test if gene-specific methylation contributes to the immortalized and transformed phenotype, we analyzed the methylation status of 17 genes in normal cells immortalized with SV40, hTERT, Ad5, Ad12-SV40 or HPV-18. Some of these immortalized lines were progressively transformed and tumorigenic in nude mice. We observed gene-specific methylation in the in vitro immortalized and transformed cells. SV40 and HPV18 immortalization resulted in different methylation spectra. In SV40- and h-TERT-immortalized prostate epithelial cells, the most frequently methylated gene was RASSF1A, while in HPV18-immortalized cell lines, the RAR-beta2 gene was universally methylated. Immortalization with SV40 resulted in methylation of a greater number of genes than immortalization with HPV. Furthermore, in SV40-immortalized cell lines, methylation affected different genes in fibroblasts compared with epithelial cells, suggesting that different mechanisms may be used by SV40 to immortalize cell lines of different origins. In HPV18-immortalized and subsequently transformed cell lines, the most commonly methylated genes were hormone responsive genes, such as AR, ER-beta and RAR-beta2. In general, more genes were methylated in neoplastically-transformed cell lines than in only immortalized cell lines, indicating that accumulation of epigenetic abnormalities may contribute to oncogenesis.

    View details for Web of Science ID 000226104600032

    View details for PubMedID 15586250

  • Interaction of nuclear receptor ligands with the vitamin D signaling pathway in prostate cancer JOURNAL OF STEROID BIOCHEMISTRY AND MOLECULAR BIOLOGY Peehl, D. M., Feldman, D. 2004; 92 (4): 307-315

    Abstract

    A number of hormonal ligands and/or the nuclear receptors that mediate their actions have been targeted for prostate cancer therapy. Androgens, the ligands for the androgen receptor (AR), are critical for the growth of prostate cancer. Inhibition of androgen production has been the mainstay of treatment for advanced prostate cancer for decades. Other more recently tested targets include retinoid receptors (RAR and RXR), glucocorticoid receptors (GR), estrogen receptors (ER) and peroxisome proliferator-activated receptors (PPAR). Calcitriol, acting through the Vitamin D receptor (VDR), has many tumor suppressive activities in the prostate, including inhibition of proliferation, induction of apoptosis and/or differentiation, and reduction of cellular invasion. Because of these properties, calcitriol and its less hypercalcemic analogs are being evaluated as agents to prevent or treat prostate cancer. Androgens, retinoids, glucocorticoids, estrogens and agonists of PPAR directly or indirectly impact Vitamin D signaling pathways, and vice versa. In order to design the most effective strategies to use calcitriol to prevent or treat prostate cancer, the interactions of other nuclear receptors and their ligands with the Vitamin D signaling pathway need to be considered.

    View details for DOI 10.1016/j.jsbmb.2004.10.006

    View details for Web of Science ID 000227022500010

    View details for PubMedID 15663994

  • Expression of transforming growth factor-beta 1 and growth in soft agar differentiate prostate carcinoma-associated fibroblasts from normal prostate fibroblasts INTERNATIONAL JOURNAL OF CANCER Francisco, I. F., DeWolf, W. C., Peehl, D. M., Olumi, A. F. 2004; 112 (2): 213-218

    Abstract

    Carcinoma-associated fibroblasts (CAF) promote tumor progression of pre-neoplastic epithelial cells. To investigate the basis of this phenomenon, we compared the properties of fibroblasts cultured from normal human prostate (NHPF) to prostate CAF. NHPF and CAF were assayed for growth potential, cell death and proliferative capacity by measuring population doubling time, cell cycle distribution and capability to form colonies in soft agar. Resistance to genotoxic (UV radiation: 0-50 J/cm2) and chemotoxic (0-200 nM Taxol) agents were compared between CAF and NHPF by measuring cell viability and cell cycle analysis. Transforming growth factor beta1 (TGF-beta1) immunoreactivity was assessed in non-malignant and malignant prostatic tissue. No detectable differences were found when comparing CAF and NHPF with respect to population doubling time, cell cycle distribution and response to genotoxic and chemotoxic agents. The mean number of colonies in soft agar was 120.5 for CAF vs. 18.2 for NHPF (p < 0.05). Because TGF-beta1 and matrix metalloproteinase (MMP)-9 have been associated with growth of fibroblasts in soft agar and tumor promotion, we measured the expression of these factors in NHPF and CAF by ELISA. There was no difference in expression of MMP-9; however, TGF-beta1 was expressed in higher concentrations in CAF than in NHPF (p < 0.0014). Furthermore, TGF-beta1 expression was higher in the carcinoma-associated stroma of prostate cancer tissue than stroma of non-malignant prostatic tissue. Increased capability of CAF as compared to NHPF to form colonies in soft agar may be due to a higher expression of TGF-beta1 and correlates with the ability of CAF to promote malignant progression of prostate epithelial cells.

    View details for DOI 10.1002/ijc.20388

    View details for Web of Science ID 000223939500006

    View details for PubMedID 15352032

  • Molecular and cellular pathogenesis of benign prostatic hyperplasia JOURNAL OF UROLOGY Lee, K. L., Peehl, D. M. 2004; 172 (5): 1784-1791

    Abstract

    Symptomatic benign prostatic hyperplasia (BPH) is one of the most common ailments seen by the urologist. Significant advances have occurred in medical and surgical therapy, and in the understanding of the biology of this disease. However, the basic science literature is often conflicting and confusing, without a unified voice. We report the current state of knowledge of the molecular and cellular basis of BPH.We compiled and interpreted basic science studies relevant to BPH pathogenesis.Cellular alterations that include changes in proliferation, differentiation, apoptosis and senescence in the epithelium and stroma are implicated in BPH pathogenesis. Molecular analyses have yielded numerous candidate genes important in disease progression. Differential expression of cytokines and growth factors in BPH tissue suggests roles for inflammation and hypoxia. Through the use of cell culture models the complex regulatory mechanisms of growth control in BPH are becoming defined.The scientific endeavor has resulted in great strides in our understanding of BPH on a molecular and cellular level. It is hopeful that basic science and translational research will improve treatment and prevention strategies for this common disease of elderly men.

    View details for DOI 10.1097/01.ju.0000133655.71782.14

    View details for Web of Science ID 000224463600005

    View details for PubMedID 15540721

  • Re-evaluation of inhibin at subunit as a tumour suppressor in prostate cancer International Workshop on Inhibins, Activins and Follistatins Risbridger, G. P., Ball, E. M., Wang, H., Mellor, S. L., Peehl, D. M. ELSEVIER IRELAND LTD. 2004: 73–76

    Abstract

    Inhibin is a member of the TGF-beta superfamily of growth and differentiation factors and a tumor suppressor. Consistent with the tumor suppressive function of the inhibin alpha subunit in prostate cancer, we reported a loss of gene expression due to DNA hypermethylation and loss of heterozygosity (LOH) as well as down regulation of inhibin alpha subunit immunoreactivity. Paradoxically, an expanded study to evaluate the prognostic significance of inhibin alpha subunit expression in men with prostate cancer resulted in a contradictory outcome, whereby an up-regulation of subunit expression was recorded. In seeking a more comprehensive explanation for all data sets, we offer a unifying hypothesis. We propose that the tumor suppressor activities of the inhibin alpha subunit dominate in non-malignant tissue, but its oncogenic activities emerge during tumorigenesis. An explanation such as this, involving a switch in gene function from being tumor suppressive to pro-oncogenic, may account for the discrepant findings in other types of cancer.

    View details for DOI 10.1016/j.mce.2004.02.015

    View details for Web of Science ID 000224623600011

    View details for PubMedID 15451570

  • Molecular activity of 1,25-dihydroxyvitamin D-3 in primary cultures of human prostatic epithelial cells revealed by cDNA microarray analysis JOURNAL OF STEROID BIOCHEMISTRY AND MOLECULAR BIOLOGY Peehl, D. M., Shinghal, R., Nonn, L., Seto, E., Krishnan, A. V., Brooks, J. D., Feldman, D. 2004; 92 (3): 131-141

    Abstract

    1,25-Dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] exerts anti-proliferative, differentiating and apoptotic effects on prostatic cells. These activities, in addition to epidemiologic findings that link Vitamin D to prostate cancer risk, support the use of 1,25(OH)(2)D(3) for prevention or therapy of prostate cancer. The molecular mechanisms by which 1,25(OH)(2)D(3) exerts antitumor effects on prostatic cells are not well-defined. In addition, there is heterogeneity among the responses of various prostate cell lines and primary cultures to 1,25(OH)(2)D(3) with regard to growth inhibition, differentiation and apoptosis. To understand the basis of these differential responses and to develop a better model of Vitamin D action in the prostate, we performed cDNA microarray analyses of primary cultures of normal and malignant human prostatic epithelial cells, treated with 50 nM of 1,25(OH)(2)D(3) for 6 and 24 h. CYP24 (25-hydroxyvitamin D(3)-24-hydroxylase) was the most highly upregulated gene. Significant and early upregulation of dual specificity phosphatase 10 (DUSP10), validated in five additional primary cultures, points to inhibition of members of the mitogen-activated protein kinase (MAPK) superfamily as a key event mediating activity of 1,25(OH)(2)D(3) in prostatic epithelial cells. The functions of other regulated genes suggest protection by 1,25(OH)(2)D(3) from oxidative stress. Overall, these results provide new insights into the molecular basis of antitumor activities of Vitamin D in prostate cells.

    View details for DOI 10.1016/j.jsbmb.2004.07.003

    View details for Web of Science ID 000226009000003

    View details for PubMedID 15555907

  • Altered SMRT levels disrupt vitamin D-3 receptor signalling in prostate cancer cells ONCOGENE Khanim, F. L., Gommersall, L. M., Wood, V. H., Smith, K. L., Montalvo, L., O'Neill, L. P., Xu, Y., Peehl, D. M., Stewart, P. M., Turner, B. M., Campbell, M. J. 2004; 23 (40): 6712-6725

    Abstract

    We hypothesized that key antiproliferative target genes for the vitamin D receptor (VDR) were repressed by an epigenetic mechanism in prostate cancer cells resulting in apparent hormonal insensitivity. To explore this possibility, we examined nuclear receptor corepressor expression in a panel of nonmalignant and malignant cell lines and primary cultures, and found frequently elevated SMRT corepressor mRNA expression often associated with reduced sensitivity to 1alpha,25-dihydroxyvitamin D(3) (1alpha,25(OH)2D3). For example, PC-3 and DU-145 prostate cancer cell lines had 1.8-fold and twofold increases in SMRT mRNA relative to normal PrEC cells (P<0.05). Similarly, 10/15 primary tumour cultures (including three matched to normal cells from the same donors) had elevated SMRT mRNA levels; generally NCoR1 and Alien were not as commonly elevated. Corepressor proteins often have associated histone deacetylases (HDAC) and reflectively the antiproliferative action of 1alpha,25(OH)2D3 can be 'restored' by cotreatment with low doses of HDAC inhibitors such as trichostatin A (TSA, 15 nM) to induce apoptosis in prostate cancer cell lines. To decipher the transcriptional events that lead to these cellular responses, we undertook gene expression studies in PC-3 cells after cotreatment of 1alpha,25(OH)2D3 plus TSA after 6 h. Examination of known VDR target genes and cDNA microarray analyses revealed cotreatment of 1alpha,25(OH)2D3 plus TSA cooperatively upregulated eight (out of 1176) genes, including MAPK-APK2 and GADD45alpha. MRNA and protein time courses and inhibitor studies confirmed these patterns of regulation. Subsequently, we knocked down SMRT levels in PC-3 cells using a small interfering RNA (siRNA) approach and found that GADD45alpha induction by 1alpha,25(OH)2D3 alone became very significantly enhanced. The same distortion of gene responsiveness, with repressed induction of GADD45alpha was found in primary tumour cultures compared and to matched peripheral zone (normal) cultures from the same donor. These data demonstrate that elevated SMRT levels are common in prostate cancer cells, resulting in suppression of target genes associated with antiproliferative action and apparent 1alpha,25(OH)2D3-insensitivity. This can be targeted therapeutically by combination treatments with HDAC inhibitors.

    View details for DOI 10.1038/sj.onc.1207772

    View details for Web of Science ID 000223653600003

    View details for PubMedID 15300237

  • CXCR4 and CXCL12 (SDF-1) in prostate cancer: Inhibitory effects of human single chain Fv antibodies CLINICAL CANCER RESEARCH Vaday, G. G., Hua, S. B., Peehl, D. M., Pauling, M. H., Lin, Y. H., Zhu, L., Lawrence, D. M., Foda, H. D., Zucker, S. 2004; 10 (16): 5630–39

    Abstract

    Metastasis is a major cause of morbidity in prostate cancer (PCa). Several studies have shown that the chemokine receptor CXCR4 and its ligand, CXCL12 (stromal cell-derived factor-1), regulate tumor cell metastasis to specific organs. Recently, it was demonstrated that CXCL12 enhances PCa cell adhesion, migration, and invasion, implicating CXCR4 in PCa metastasis. In this study, we examined the inhibitory effects of anti-CXCR4 antibodies on CXCL12-mediated PCa cell activities.We developed fully human single chain Fv antibodies (scFv), Ab124 and Ab125, against CXCR4 using the yeast two-hybrid system. We performed immunofluorescent staining, flow cytometry, and ELISA-binding assays to measure scFv binding to PCa cells. We also examined the effects of scFv on CXCL12-mediated calcium mobilization, cell migration, and invasion.Our results confirmed that PCa cell lines express cell-surface CXCR4. Real-time quantitative reverse transcription-PCR and immunohistochemical staining also verified that CXCR4 is expressed in primary cultures of prostate epithelial cells from adenocarcinomas and in human prostate tissues. Ab124 and Ab125 demonstrated specific binding to PCa cell lines by flow cytometry and in binding assays. Preincubation with scFv resulted in significant reduction of CXCL12-induced calcium mobilization in PC-3 and LNCaP cells. Ab124 and Ab125 also inhibited PCa cell migration toward CXCL12, as well as invasion through extracellular matrix gels.Ab124 and Ab125 inhibit CXCL12-mediated cellular activities by binding the receptor CXCR4. Recombinant scFv are an efficient mode of targeting tumor antigens. Considering the high incidence of PCa, the development of fully human scFv may be a useful therapeutic approach in the prevention and treatment of PCa metastasis.

    View details for DOI 10.1158/1078-0432.CCR-03-0633

    View details for Web of Science ID 000223454600043

    View details for PubMedID 15328206

  • Targeting SMRT co-repressor complexes as a novel chemoprevention strategy for prostate cancer British Cancer Research Meeting 2004 Gommersall, L. M., Khanim, F. L., Peehl, D. M., Doherty, A. P., James, N. D., Campbell, M. J. NATURE PUBLISHING GROUP. 2004: S20–S20
  • Mechanisms of decreased Vitamin D 1 alpha-hydroxylase activity in prostate cancer cells MOLECULAR AND CELLULAR ENDOCRINOLOGY Ma, J. F., Nonn, L., Campbell, M. J., Hewison, M., Feldman, D., Peehl, D. M. 2004; 221 (1-2): 67-74

    Abstract

    Vitamin D 1alpha-hydroxylase (1alpha(OH)ase), which converts the circulating prohormone 25-hydroxyvitamin-D(3) (25(OH)D(3)) to the active 1alpha-25-dihydroxyvitamin-D(3) (1,25(OH)(2)D(3)), is present in normal prostatic epithelium. However, prostate cancer cells, both primary cultured cells and cell lines, have greatly decreased activity of 1alpha(OH)ase and are therefore resistant to the tumor suppressor activity of circulating 25(OH)D(3). We quantitated 1alpha(OH)ase mRNA and protein levels to investigate mechanism(s) responsible for decreased 1alpha(OH)ase enzymatic activity in prostate cancer. Prostate cancer cell lines had low 1alpha(OH)ase mRNA levels. Primary prostate cell cultures derived from normal and cancer tissues had equivalent levels of 1alpha(OH)ase RNA and protein. Equivalent 1alpha(OH)ase protein levels were observed in prostate tissue sections containing normal and malignant cells. The protein levels of hsc70, whose homolog intracellular Vitamin D binding protein (IDBP-1) facilitates delivery of 25(OH)D(3) to 1alpha(OH)ase in monkey cells, were equivalent in the normal and cancer cells. Equivalent activity in normal and cancer cells of Vitamin D 24-hydroxylase, a mitochondrial enzyme that also uses 25(OH)D(3) as a substrate, further ruled out lack of access to substrate as a basis for low activity of 1alpha(OH)ase in cancer cells. We conclude that diminished 1alpha(OH)ase activity in prostate cancer cell lines is through decreased gene expression, whereas decreased activity in primary cultures and tissues is post-translational.

    View details for DOI 10.1016/j.mce.2004.03.003

    View details for Web of Science ID 000222854100007

    View details for PubMedID 15223133

  • Analysis of vitamin D-regulated gene expression in LNCaP human prostate cancer cells using cDNA microarrays PROSTATE Krishnan, A. V., Shinghal, R., Raghavachari, N., Brooks, J. D., Peehl, D. M., Feldman, D. 2004; 59 (3): 243-251

    Abstract

    1,25-dihydroxyvitamin D(3) [1,25(OH)2D3] exerts growth inhibitory, pro-differentiating, and pro-apoptotic effects on prostate cells. To better understand the molecular mechanisms underlying these actions, we employed cDNA microarrays to study 1,25(OH)2D3-regulated gene expression in the LNCaP human prostate cancer cells.mRNA isolated from LNCaP cells treated with vehicle or 50 nM 1,25(OH)2D3 for various lengths of time were hybridized to microarrays carrying approximately 23,000 genes. Some of the putative target genes revealed by the microarray analysis were verified by real-time PCR assays.1,25(OH)2D3 most substantially increased the expression of the insulin-like growth factor binding protein-3 (IGFBP-3) gene. Our analysis also revealed several novel 1,25(OH)2D3-responsive genes. Interestingly, some of the key genes regulated by 1,25(OH)2D3 are also androgen-responsive genes. 1,25(OH)2D3 also down-regulated genes that mediate androgen catabolism.The putative 1,25(OH)2D3 target genes appear to be involved in a variety of cellular functions including growth regulation, differentiation, membrane transport, cell-cell and cell-matrix interactions, DNA repair, and inhibition of metastasis. The up-regulation of IGFBP-3 gene has been shown to be crucial in 1,25(OH)2D3-mediated inhibition of LNCaP cell growth. 1,25(OH)2D3 regulation of androgen-responsive genes as well as genes involved in androgen catabolism suggests that there are interactions between 1,25(OH)2D3 and androgen signaling pathways in LNCaP cells. Further studies on the role of these genes and others in mediating the anti-cancer effects of 1,25(OH)2D3 may lead to better approaches to the prevention and treatment of prostate cancer.

    View details for DOI 10.1002/pros.20006

    View details for Web of Science ID 000220910600003

    View details for PubMedID 15042599

  • Epigenetic repression of transcription by the vitamin D-3 receptor in prostate cancer cells JOURNAL OF STEROID BIOCHEMISTRY AND MOLECULAR BIOLOGY Gommersall, L. M., Khanim, F. L., Peehl, D. M., Doherty, A. H., Campbell, M. J. 2004; 89-90 (1-5): 251-256

    Abstract

    Normal prostate epithelial cells are acutely sensitive to the antiproliferative action of 1alpha,25-dihydroxyvitamin D(3) (1alpha,25(OH)(2)D(3)), whilst prostate cancer cell lines and primary cultures display a range of sensitivities. We hypothesised that key antiproliferative target genes of the Vitamin D receptor (VDR) were repressed by an epigenetic mechanism in 1alpha,25(OH)(2)D(3)-insensitive cells. Supportively, we found elevated nuclear receptor co-repressor and reduced VDR expression correlated with reduced sensitivity to the antiproliferative action of 1alpha,25(OH)(2)D(3). Furthermore, the growth suppressive actions of 1alpha,25(OH)(2)D(3) can be restored by co-treatment with low doses of histone deacetylation inhibitors, such as trichostatin A (TSA) to induce apoptosis. Examination of the regulation of VDR target genes revealed that co-treatment of 1alpha,25(OH)(2)D(3) plus TSA co-operatively upregulated GADD45alpha. Similarly in a primary cancer cell culture, the regulation of appeared GADD45alpha repressed. These data demonstrate that prostate cancer cells utilise a mechanism involving deacetylation to suppress the responsiveness of VDR target genes and thus ablate the antiproliferative action of 1alpha,25(OH)(2)D(3).

    View details for DOI 10.1016/j.jsbmb.2004.03.080

    View details for Web of Science ID 000222852500044

    View details for PubMedID 15225780

  • Are primary cultures realistic models of prostate cancer? JOURNAL OF CELLULAR BIOCHEMISTRY Peehl, D. M. 2004; 91 (1): 185-195

    Abstract

    Primary cultures fill a unique niche among the repertoire of in vitro model systems available to investigate the biology of the normal and malignant human prostate. This review summarizes some of the properties of primary cultures, with special emphasis on two questions: are primary cultures from adenocarcinomas really comprised of cancer rather than normal cells, and do primary cultures faithfully retain characteristics of cells of origin?

    View details for DOI 10.1002/jcb.10691

    View details for Web of Science ID 000187714700016

    View details for PubMedID 14689590

  • Elevated expression of inhibin alpha in prostate cancer JOURNAL OF UROLOGY Risbridger, G. P., Shibata, A., Ferguson, K. L., Stamey, T. A., McNeal, J. E., Peehl, D. M. 2004; 171 (1): 192-196

    Abstract

    Less than 50% of men who undergo radical prostatectomy for prostate cancer are cured of disease. We evaluate tumor expression of inhibin alpha, a putative tumor suppressor, and the related protein, follistatin, to determine whether expression correlated with failure to be cured by surgery.Tissues were selected from an archival collection of 379 prostatectomy specimens from men with followup of at least 5 years after surgery. Since previous studies showed that such men with only Gleason grade 3 cancer had a greater than 95% chance of no biochemical recurrence (increase in serum prostate specific antigen), our investigation was confined to 174 men with 2% or greater grade 4/5 cancer. These men had an intermediate rate of failure, providing an opportunity to analyze the potential contribution of inhibin alpha or follistatin to progression. Intensity of immunohistochemical labeling for inhibin alpha and follistatin in each cancer was compared with that in normal glands within the same tissue section.The majority of cases showed more intense expression of inhibin alpha in cancer than in normal glands. Those individuals whose cancers had the most elevated expression of inhibin alpha had a higher risk of recurrence, although this association was not statistically significant. Follistatin was expressed equivalently in normal and cancer cells in the majority of cases and did not correlate with recurrence.Our finding that inhibin alpha is frequently overexpressed in high grade prostate cancer suggests that the role of inhibin alpha as a tumor suppressor needs to be reevaluated. Furthermore, assessment of inhibin alpha as a serum marker of prostate cancer, as used to diagnose ovarian cancer, may be warranted.

    View details for DOI 10.1097/01.ju.0000100048.98807.b7

    View details for Web of Science ID 000187390800044

    View details for PubMedID 14665874

  • Oxidative 3 alpha-hydroxysteroid dehydrogenase activity of human type 10 17 beta-hydroxysteroid dehydrogenase JOURNAL OF STEROID BIOCHEMISTRY AND MOLECULAR BIOLOGY He, X. Y., Yang, Y. Z., Peehl, D. M., Lauderdale, A., SCHULZ, H., Yang, S. Y. 2003; 87 (2-3): 191-198

    Abstract

    In vitro enzyme assays have demonstrated that human type 10 17beta-hydroxysteroid dehydrogenase (17beta-HSD10) catalyzes the oxidation of 5alpha-androstane-3alpha,17beta-diol (adiol), an almost inactive androgen, to dihydrotestosterone (DHT) rather than androsterone or androstanedione. To further investigate the role of this steroid-metabolizing enzyme in intact cells, we produced stable transfectants expressing 17beta-HSD10 or its catalytically inactive Y168F mutant in human embryonic kidney (HEK) 293 cells. It was found that DHT levels in HEK 293 cells expressing 17beta-HSD10, but not its catalytically inactive mutant, will dramatically increase if adiol is added to culture media. Moreover, certain malignant prostatic epithelial cells have more 17beta-HSD10 than normal controls, and can generate DHT, the most potent androgen, from adiol. This event might promote prostate cancer growth. Analysis of the 17beta-HSD10 sequence shows that this enzyme does not have any ER retention signal or transmembrane segments and has not originated by divergence from a retinol dehydrogenase. The data suggest that the unique mitochondrial location of this HSD [Eur. J. Biochem. 268 (2001) 4899] does not prevent it from oxidizing the 3alpha-hydroxyl group of a C19 sterol in living cells. The experimental results lead to the conclusion that mitochondrial 17beta-HSD10 plays a significant part in a non-classical androgen synthesis pathway along with microsomal retinol dehydrogenases.

    View details for DOI 10.1016/j.jsbmb.2003.07.007

    View details for Web of Science ID 000188091500009

    View details for PubMedID 14672739

  • Elevated expression of inhibitor of apoptosis proteins in prostate cancer CLINICAL CANCER RESEARCH Krajewska, M., Krajewski, S., Banares, S., Huang, X. S., Turner, B., Bubendorf, L., Kallioniemi, O. P., Shabaik, A., Vitiello, A., Peehl, D., Gao, G. J., Reed, J. C. 2003; 9 (13): 4914-4925

    Abstract

    Inhibitor of apoptosis (IAP) family proteins are suppressors of apoptosis that have been implicated in apoptosis resistance in some cancers. Their expression and relevance to the prognosis of prostate cancer were investigated.The expression of four members of the IAP family (cellular inhibitor of apoptosis protein 1, cellular inhibitor of apoptosis protein 2, X chromosome-linked IAP, and survivin) was examined by immunohistochemistry and immunoblotting in human prostate cancers and in prostate tissues from transgenic mice expressing SV40 large T antigen under control of a probasin promoter.Tumor-associated elevations in the levels of all four IAP family members were common in prostate cancers of both humans and mice, suggesting concomitant up-regulation of multiple IAP family proteins. Compared with normal prostatic epithelium, increased IAP expression was often evident even in prostatic intraepithelial neoplasia lesions (carcinoma in situ), suggesting that deregulation of IAP expression occurs early in the pathogenesis of prostate cancer. IAP expression did not correlate with Gleason grade or prostate-specific antigen levels.The findings demonstrate that tumor- associated elevations in the expression of several IAP family proteins occur as a frequent and early event in the etiology of prostate cancer.

    View details for Web of Science ID 000186212000034

    View details for PubMedID 14581366

  • Pathways mediating the growth-inhibitory actions of vitamin D in prostate cancer Conference on Nutritional Genomics and Proteomics in Cancer Prevention Peehl, D. M., Krishnan, A. V., Feldman, D. AMER SOC NUTRITIONAL SCIENCE. 2003: 2461S–2469S

    Abstract

    Vitamin D is emerging as an important dietary factor that affects the incidence and progression of many malignancies including prostate cancer. The active form of vitamin D, 1,25-dihydroxycholecalciferol [1,25(OH)(2)D(3)], inhibits the growth and stimulates the differentiation of prostate cancer cells. We have studied primary cultures of normal and cancer-derived prostatic epithelial cells as well as established human prostate cancer cell lines to elucidate the molecular pathways of 1,25(OH)(2)D(3) actions. These pathways are varied and appear to be cell specific. In LNCaP cells, 1,25(OH)(2)D(3) mainly causes growth arrest through the induction of insulin-like growth factor binding protein-3 and also stimulates apoptosis to a much smaller extent. We have used cDNA-microarray analyses to identify additional genes that are regulated by 1,25(OH)(2)D(3) and to raise novel therapeutic targets for use in the chemoprevention or treatment of prostate cancer. Less calcemic analogs of 1,25(OH)(2)D(3) that have more antiproliferative activity are being developed that will be more useful clinically. In target cells, 1,25(OH)(2)D(3) induces 24-hydroxylase, the enzyme that catalyzes its self inactivation. Cotreatment with 24-hydroxylase inhibitors enhances the antiproliferative activity of 1,25(OH)(2)D(3). The combination of other anticancer agents such as retinoids with vitamin D offers another promising therapeutic approach. A small clinical trial has shown that 1,25(OH)(2)D(3) can slow the rate of prostate-specific antigen increase in prostate cancer patients, which demonstrates proof of the concept that vitamin D or its analogs are clinically effective. Our research is directed at understanding the mechanisms of vitamin D action in prostate cells with the goal of developing chemoprevention and treatment strategies to improve prostate cancer therapy.

    View details for Web of Science ID 000184163200012

    View details for PubMedID 12840225

  • Primary culture model of peroxisome proliferator-activated receptor gamma activity in prostate cancer cells JOURNAL OF CELLULAR PHYSIOLOGY Xu, Y., Iyengar, S., Roberts, R. L., Shappell, S. B., Peehl, D. M. 2003; 196 (1): 131-143

    Abstract

    BRL 49653 (rosiglitazone) is a thiazolidinedione anti-diabetic drug that activates the nuclear receptor, peroxisome proliferator-activated receptor gamma (PPARgamma). Pilot clinical trials have shown evidence of therapeutic activity of PPARgamma agonists against prostate cancer. To more effectively use PPARgamma ligands to treat this common and generally chemo-resistant type of cancer, it will be necessary to better understand the nature of PPARgamma activity in prostate cancer cells. Tumor suppressor effects of activation of PPARgamma may include suppression of growth and/or induction of differentiation or apoptosis. We investigated responses of primary cultures of human prostatic cancer cells to BRL 49653. PPARgamma was expressed in all of the cell strains examined. BRL 49653 caused dose- and time-dependent growth inhibition that was associated with increased expression of the transcription repressor, transforming growth factor beta-stimulated clone 22 (TSC-22), and markedly increased expression of the secretory differentiation-associated gene adipophilin. Adipocyte-type fatty acid binding protein (aFABP), neutrophil gelatinase-associated lipocalin (NGAL), glycerol kinase (GyK), and beta-catenin, which are regulated by PPARgamma ligands in certain other types of cells, were not regulated by BRL 49653 in prostate cells. Upregulation of adipophilin coincided with morphological changes and the appearance of cytoplasmic vacuoles with ultrastructural features of secondary lysosomes. These results extend previous studies with established cancer cell lines and show that PPARgamma agonists can inhibit proliferation and modulate expression of secretory-associated genes in primary cultures of prostate cancer cells, further warranting consideration of these agents as pro-differentiating chemotherapeutic or chemoprevention agents for the treatment of prostate cancer.

    View details for DOI 10.1002/jcp.10281

    View details for PubMedID 12767049

  • Human type 3 3 alpha-hydroxysteroid dehydrogenase (Aldo-Keto reductase 1C2) and androgen metabolism in prostate cells ENDOCRINOLOGY Rizner, T. L., Lin, H. K., Peehl, D. M., Steckelbroeck, S., Bauman, D. R., Penning, T. M. 2003; 144 (7): 2922-2932

    Abstract

    Human aldo-keto reductases (AKRs) of the AKR1C subfamily function in vitro as 3-keto-, 17-keto-, and 20-ketosteroid reductases or as 3alpha-, 17beta-, and 20alpha-hydroxysteroid oxidases. These AKRs can convert potent sex hormones (androgens, estrogens, and progestins) into their cognate inactive metabolites or vice versa. By controlling local ligand concentration AKRs may regulate steroid hormone action at the prereceptor level. AKR1C2 is expressed in prostate, and in vitro it will catalyze the nicotinamide adenine dinucleotide (NAD(+))-dependent oxidation of 3alpha-androstanediol (3alpha-diol) to 5alpha-dihydrotestosterone (5alpha-DHT). This reaction is potently inhibited by reduced NAD phosphate (NADPH), indicating that the NAD(+): NADPH ratio in cells will determine whether AKR1C2 makes 5alpha-DHT. In transient COS-1-AKR1C2 and in stable PC-3-AKR1C2 transfectants, 5alpha-DHT was reduced by AKR1C2. However, the transfected AKR1C2 oxidase activity was insufficient to surmount the endogenous 17beta-hydroxysteroid dehydrogenase (17beta-HSD) activity, which eliminated 3alpha-diol as androsterone. PC-3 cells expressed retinol dehydrogenase/3alpha-HSD and 11-cis-retinol dehydrogenase, but these endogenous enzymes did not oxidize 3alpha-diol to 5alpha-DHT. In stable LNCaP-AKR1C2 transfectants, AKR1C2 did not alter androgen metabolism due to a high rate of glucuronidation. In primary cultures of epithelial cells, high levels of AKR1C2 transcripts were detected in prostate cancer, but not in cells from normal prostate. Thus, in prostate cells AKR1C2 acts as a 3-ketosteroid reductase to eliminate 5alpha-DHT and prevents activation of the androgen receptor. AKR1C2 does not act as an oxidase due to either potent product inhibition by NADPH or because it cannot surmount the oxidative 17beta-HSD present. Neither AKR1C2, retinol dehydrogenase/3alpha-HSD nor 11-cis-retinol dehydrogenase is a source of 5alpha-DHT in PC-3 cells.

    View details for DOI 10.1210/en.2002-0032

    View details for Web of Science ID 000183547800024

    View details for PubMedID 12810547

  • The role of vitamin D and retinoids in controlling prostate cancer progression 11th International Congress on Hormonal Steroids/7th International Congress on Hormones and Cancer Peehl, D. M., Feldman, D. BIOSCIENTIFICA LTD. 2003: 131–40

    Abstract

    Prostate cancer is a leading cause of cancer-related deaths in many countries. Premalignant lesions and invasive cancer occur more frequently in the prostate than in any organ other than the skin. Yet, the incidence of clinically detected prostate cancer is much lower than the histopathological incidence. The slow growth of prostate cancer and the low incidence of clinically manifest disease in some geographical locations or racial/ethnic groups suggest that prostate cancer can be controlled, perhaps by dietary factors. Vitamin D and retinoids have emerged as leading candidates both to prevent and to treat prostate cancer. Many of the activities of these compounds, established from epidemiological studies, research with cell culture and animal models, and clinical trials, are consistent with tumor suppressor effects. However, retinoids may have additional tumor enhancer properties that balance or negate anti-cancer activity. This perhaps explains the overall lack of protective effects of vitamin A compounds against prostate cancer found in epidemiological studies, and the minimal efficacy of retinoids in clinical trials to treat prostate cancer. While current efforts focus on developing strategies to use vitamin D compounds to control prostate cancer, the possibility exists that prostate cancer cells may become resistant to tumor suppressor effects of vitamin D. Analyses of experimental model systems show that prostate cancer cells become less sensitive to vitamin D through loss of receptors or signaling molecules that mediate vitamin D's actions, or through changes in metabolic enzymes that synthesize or degrade vitamin D compounds. The potential promise of exploiting vitamin D to control prostate cancer is tempered by the possibility that prostate cancer, perhaps even at early stages, may develop mechanisms to escape tumor suppressor activities of vitamin D and/or retinoids.

    View details for Web of Science ID 000184151400003

    View details for PubMedID 12790775

  • Novel stimulatory role for insulin-like growth factor binding protein-2 in prostate cancer cells INTERNATIONAL JOURNAL OF CANCER Moore, M. G., Wetterau, L. A., Francis, M. J., Peehl, D. M., Cohen, P. 2003; 105 (1): 14-19

    Abstract

    The insulin-like growth factor (IGF) axis is a complex system composed of 2 mitogenic ligands, IGF-I and -II, 2 receptors, IGF-1R and IGF-2R, and 6 binding proteins, IGFBP-1 to -6. The IGFBPs exert their actions through their regulation of IGF bioavailability for IGF receptors. In addition, some IGFBPs have also been found to have direct cellular actions independent of IGFs. IGFBP-2 is a major IGFBP in the prostate and in seminal fluid. IGFBP-2 levels, which are elevated in many malignancies, are markedly increased in prostate cancer, and show a positive correlation with prostate specific antigen (PSA) and prostate tumor aggressiveness. We investigated the potential role of IGFBP-2 in the pathogenesis of prostate cancer by evaluating its ability to stimulate growth and the expression and activity of the nuclear enzyme, telomerase. We found IGFBP-2 to have a modest suppressive effect on the growth of primary cultures of normal prostate epithelial cells and a potent IGF-antagonistic effect in these cells, similar to previous reports on the inhibitory nature of this molecule. Surprisingly, IGFBP-2 had a potent stimulatory effect on growth of LAPC-4 prostate cancer cells, an effect that was more pronounced in the absence of androgens. IGFBP-2 growth stimulation of LAPC-4 cells was completely blocked by MAP-kinase inhibitors and partially blocked by PI3-kinase inhibitors. IGFBP-2 stimulation of LAPC-4 cell growth seen in serum-free conditions was lost in the presence of 10% FBS. IGFBP-2 also had a growth stimulatory effect on DU 145 prostate cancer cells. IGFBP-2 significantly stimulated telomerase activity in LAPC-4 cells in the absence of androgens. In addition, IGFBP-2 significantly stimulated hTERT expression and telomerase activity in DU 145 cells. Thus, we demonstrated an inhibitory effect of IGFBP-2 on non-malignant prostate cells, but showed it to be stimulatory for prostate cancer cells in a MAP-kinase and androgen-modulated process. In conclusion, IGFBP-2 may play a role in the progression, but not in the initiation of the prostate cancer disease process, suggesting the existence of a molecular switch transitioning IGFBP-2 from a growth inhibitor to a pro-carcinogenic molecule.

    View details for DOI 10.1002/ijc.11015

    View details for Web of Science ID 000182163000003

    View details for PubMedID 12672024

  • Insulin-like Growth Factor Binding Protein-3 (IGFBP-3) induces apoptosis via rapid RXR alpha-dependent TR3 mitochondrial targeting in cancer cells Lee, K. W., Ma, L. Q., Liu, B. R., Peehl, D. M., Zhang, X. K., Cohen, P. INT PEDIATRIC RESEARCH FOUNDATION, INC. 2003: 154A
  • Leptomycin B stabilizes and activates p53 in primary prostatic epithelial cells and induces apoptosis in the LNCaP cell line PROSTATE Lecane, P. S., Kiviharju, T. M., Sellers, R. G., Peehl, D. M. 2003; 54 (4): 258-267

    Abstract

    Previous studies showed that primary cultures of normal and malignant human prostatic epithelial cells are defective in their ability to upregulate the tumor suppressor protein p53 in response to DNA damage. This dysfunctional regulation of p53 may be relevant to both the high incidence of prostate cancer and its resistance to chemotherapy. Leptomycin B (LMB) has recently been found to increase the protein level and transcriptional activity of p53 by interfering with nucleocytoplasmic export and subsequent degradation by the proteasome. We investigated the ability of LMB to activate p53 in prostatic epithelial cells.Primary cultures and the cell lines LNCaP and DU 145 were treated with LMB. p53 protein was evaluated in Western blots and by immunocytochemistry. Induction of downstream targets of p53 was evaluated in Western and Northern blots. Growth inhibition, cell cycle arrest, and apoptosis in response to LMB were measured in clonal growth assays, by flow cytometry, and by Hoescht/propidium iodide staining, respectively.Treatment of prostatic epithelial cells with LMB led to post-translational stabilization of p53, activation of downstream target genes, and induction of cell cycle arrest in primary cultures and apoptosis in LNCaP (with wild-type p53) but not DU 145 (with mutant p53) cells.p53 in primary cultures of normal and malignant prostate cells, although dysfunctional in that it is not responsive to DNA damage, is activated by LMB. The ability of LMB to stabilize p53 and induce expression of p53-responsive growth inhibitory genes may be a useful lead in the development of chemopreventive or therapeutic small molecules that can modulate p53 function in prostatic epithelial cells.

    View details for DOI 10.1002/pros.10197

    View details for Web of Science ID 000181056800002

    View details for PubMedID 12539224

  • Inhibition of prostate cancer growth by vitamin D: Regulation of target gene expression JOURNAL OF CELLULAR BIOCHEMISTRY Krishnan, A. V., Peehl, D. M., Feldman, D. 2003; 88 (2): 363-371

    Abstract

    Prostate cancer (PCa) cells express vitamin D receptors (VDR) and 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) inhibits the growth of epithelial cells derived from normal, benign prostate hyperplasia, and PCa as well as established PCa cell lines. The growth inhibitory effects of 1,25(OH)(2)D(3) in cell cultures are modulated tissue by the presence and activities of the enzymes 25-hydroxyvitamin D(3) 24-hydroxylase which initiates the inactivation of 1,25(OH)(2)D(3) and 25-hydroxyvitamin D(3) 1alpha-hydroxylase which catalyses its synthesis. In LNCaP human PCa cells 1,25(OH)(2)D(3) exerts antiproliferative activity predominantly by cell cycle arrest through the induction of IGF binding protein-3 (IGFBP-3) expression which in turn increases the levels of the cell cycle inhibitor p21 leading to growth arrest. cDNA microarray analyses of primary prostatic epithelial and PCa cells reveal that 1,25(OH)(2)D(3) regulates many target genes expanding the possible mechanisms of its anticancer activity and raising new potential therapeutic targets. Some of these target genes are involved in growth regulation, protection from oxidative stress, and cell-cell and cell-matrix interactions. A small clinical trial has shown that 1,25(OH)(2)D(3) can slow the rate of prostate specific antigen (PSA) rise in PCa patients demonstrating proof of concept that 1,25(OH)(2)D(3) exhibits therapeutic activity in men with PCa. Further investigation of the role of calcitriol and its analogs for the therapy or chemoprevention of PCa is currently being pursued.

    View details for DOI 10.1002/jcb.10334

    View details for Web of Science ID 000180479100024

    View details for PubMedID 12520538

  • The role of vitamin D in prostate cancer 1st International Symposium on Vitamin D Analogs in Cancer Prevention and Therapy Krishnan, A. V., Peehl, D. M., Feldman, D. SPRINGER-VERLAG BERLIN. 2003: 205–221

    Abstract

    Prostate cancer (PCa) cells harbor receptors for vitamin D (VDR) as well as androgens (AR). 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] increases AR expression and enhances androgen actions linking the two receptor systems. 1,25(OH)2D3 exhibits antiproliferative activity in both AR-positive and AR-negative PCa cells. Less calcemic analogs of 1,25(OH)2D3, with more antiproliferative activity, are being developed and will be more useful clinically. The mechanisms underlying differential analog activity are being investigated. In target cells, 1,25(OH)2D3 induces 24-hydroxylase, the enzyme that catalyzes its self-inactivation. Co-treatment with 24-hydroxylase inhibitors enhances the antiproliferative activity of calcitriol. Primary cultures of normal or cancer-derived prostatic epithelial cells express 1alpha-hydroxylase, the enzyme that catalyzes the synthesis of 1,25(OH)2D3, the levels being much lower in the cancer-derived cells and in PCa cell lines. This finding raises the possibility of using 25-hydroxyvitamin D3 [25(OH)D3] as a chemopreventive agent in PCa. In LNCaP human PCa cells, 1,25(OH)2D3 and its analogs exert antiproliferative activity predominantly by cell cycle arrest, but also induce apoptosis, although to a much lesser degree. Growth arrest is mediated by induction of IGF binding protein-3 (IGFBP-3), which in turn increases the expression of the cell cycle inhibitor p21, leading to growth arrest. Other actions of 1,25(OH)2D3 in PCa cells include promotion of pro-differentiation effects and inhibition of tumor cell invasion, metastasis and angiogenesis. Combination therapy with retinoids, other anticancer agents or 24-hydroxylase inhibitors augments the inhibitory activity of 1,25(OH)2D3 in PCa and provides another effective approach in PCa treatment. Small clinical trials have shown that 1,25(OH)2D3 can slow the rate of prostate specific antigen (PSA) rise in PCa patients, demonstrating proof of concept that 1,25(OH)2D3 or its analogs will be clinically effective in PCa therapy. Current research involves further investigation of the role of 1,25(OH)2D3 and its analogs for the therapy or chemoprevention of PCa.

    View details for Web of Science ID 000184907200015

    View details for PubMedID 12899524

  • Growth of prostatic epithelial and stromal cells in vitro. Methods in molecular medicine Peehl, D. M. 2003; 81: 41-57

    View details for PubMedID 12725113

  • Vitamin D receptor start codon polymorphism (FokI) and prostate cancer progression CANCER EPIDEMIOLOGY BIOMARKERS & PREVENTION Xu, Y., Shibata, A., McNeal, J. E., Stamey, T. A., Feldman, D., Peehl, D. M. 2003; 12 (1): 23-27

    Abstract

    Vitamin D plays an important role in cell growth and differentiation and is proposed to protect against cancer initiation and/or progression. The vitamin D receptor (VDR) has a thymine/cytosine (T/C) polymorphism located in the first of two potential start (ATG) codons that can be detected by a RFLP using the endonuclease FokI. The C variant, which lacks the first ATG, results in a shorter VDR and is referred to as the F allele. The T variant (f allele) initiates at the first ATG. We examined the association of the VDR FokI genotype with histopathological characteristics and prognosis of prostate cancer among 191 mostly Caucasian subjects who had undergone radical prostatectomy between 1984 and 1992. The frequencies of the FF, Ff, and ff genotypes were 41%, 38%, and 21%, respectively. Subjects with the ff genotype had a lower mean percentage of Gleason grade 4/5 cancer (30.3%) than subjects with the FF or Ff genotypes (42.8% and 43.8%, respectively; P = 0.015 by t test for ff versus FF + Ff). The data suggest that the presence of an F allele increased the risk of being diagnosed with more aggressive cancer because higher percentage of Gleason grade 4/5 is associated with worse prognosis. The age-adjusted risk of prostate-specific antigen failure was lower for the ff genotype than for the FF genotype by Cox proportional hazards analysis but did not achieve statistical significance (hazard ratio = 0.76; 95% confidence interval, 0.44-1.32). This risk reduction disappeared after further adjustment for percentage of Gleason grade 4/5, cancer volume, and preoperative serum prostate-specific antigen level (hazard ratio = 1.03; 95% confidence interval, 0.58-1.85). In conclusion, the ff genotype was associated with less aggressive histopathological findings than Ff or FF genotypes. Additional studies with a larger sample size and investigation of the functional significance of the FokI polymorphism in prostate cancer cells are warranted.

    View details for PubMedID 12540499

  • Preclinical activity of ketoconazole in combination with calcitriol or the vitamin D analogue EB 1089 in prostate cancer cells JOURNAL OF UROLOGY Peehl, D. M., Seto, E., Hsu, J. Y., Feldman, D. 2002; 168 (4): 1583-1588

    Abstract

    Ketoconazole is a general inhibitor of P450 enzymes, of which some are necessary for androgen biosynthesis and the metabolism of vitamin D compounds. We tested the growth inhibitory activity of ketoconazole combined with 1,25-dihydroxyvitamin D3 (calcitriol) and with the vitamin D analogue EB 1089 in a preclinical model of prostate cancer.Clonal assays with primary cultures of human prostatic cancer cells were performed to test anti-proliferative effects of ketoconazole alone or in combination with calcitriol or EB 1089. Enzyme substrate reactions were done to determine whether the ability of ketoconazole to potentiate the activity of calcitriol or EB 1089 was due to the inhibition of 25-hydroxyvitamin D3-24-hydroxylase (24-hydroxylase), the enzyme that initiates conversion of active vitamin D compounds to inactive products.Ketoconazole, calcitriol and EB 1089 each inhibited the growth of prostatic cancer cells. In combination 0.1 microg./ml. ketoconazole potentiated growth inhibitory activity of calcitriol 50-fold and EB 1089 10-fold. Induction of 24-hydroxylase by calcitriol or EB 1089 was partially blocked by this level of ketoconazole.Combination therapy with ketoconazole and calcitriol or EB 1089 may enhance antitumor activities of vitamin D compounds for prostate cancer and alleviate side effects of vitamin D deficiency that are likely associated with ketoconazole therapy.

    View details for DOI 10.1097/01.ju.0000030158.18335.84

    View details for Web of Science ID 000178187800086

    View details for PubMedID 12352462

  • Polymorphisms in the androgen receptor and type II 5 alpha-reductase genes and prostate cancer prognosis PROSTATE Shibata, A., Garcia, M. I., Cheng, I., Stamey, T. A., McNeal, J. E., Brooks, J. D., Henderson, S., Yemoto, C. E., Peehl, D. M. 2002; 52 (4): 269-278

    Abstract

    Cytosine-adenine-guanine repeat length of the androgen receptor gene and the A49T and V89L polymorphisms of the 5 alpha-reductase (SRD5A2) gene have been associated with prostate cancer.We investigated the relationship of the three genetic polymorphisms to tumor grade among 211 men who had undergone radical prostatectomy. Subjects had prostate cancer <3 cm(3) with a percentage of cancer represented by Gleason grade 4 or 5 (% Gleason grade 4/5) of either > or = 20% or < or = 5%. We also examined the association between those genetic markers and prostate specific antigen (PSA) failure among 112 subjects with > or = 20% Gleason grade 4/5.In cross-sectional analysis, none of the polymorphisms was a significant predictor of % Gleason grade 4/5. In longitudinal analysis, the LL genotype at the V89L site was associated with statistically significant four- to sixfold increase in PSA failure risk after adjustment for clinicopathologic variables.We observed poorer prognosis among men with the LL genotype at codon 89 of the SRD5A2 gene. Lack of consistency between studies must be resolved before clinical utility of this marker is established.

    View details for DOI 10.1002/pros.10119

    View details for Web of Science ID 000177541200003

    View details for PubMedID 12210487

  • Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells CLINICAL CANCER RESEARCH Kiviharju, T. M., Lecane, P. S., Sellers, R. G., Peehl, D. M. 2002; 8 (8): 2666-2674

    Abstract

    Interest in exploiting traditional medicines for prevention or treatment of cancer is increasing. Extracts from the herb Tripterygium wilfordii hook F have been used in China for centuries to treat immune-related disorders. Recently it was reported that triptolide (PG490), a purified compound from Tripterygium, possessed antitumor properties and induced apoptosis by p53-independent mechanisms in a variety of malignant cell lines. This property of triptolide attracted our attention because we have found that primary cultures of human prostatic epithelial cells derived from normal tissues and adenocarcinomas are in general extremely resistant to apoptosis. Furthermore, the function of wild-type p53 is impaired in these cells such that drugs that require p53 activity to induce cell death are ineffective. Therefore, the properties of triptolide and the recent approval of its water-soluble form (PG490-88) for entry into Phase I clinical trials suggested that this drug was a promising candidate to test for antitumor activity against prostate cells. Experiments presented here demonstrated that triptolide had dose-dependent effects on both normal and cancer-derived primary cultures of human prostatic epithelial cells. Low concentrations of triptolide inhibited cell proliferation and induced a senescence-like phenotype. Higher concentrations of triptolide induced apoptosis that was unexpectedly associated with nuclear accumulation of p53. Paradoxically, levels of the p53 target genes, p21(WAF1/CIP1) and hdm-2, were reduced, as was bcl-2. Our preclinical studies suggest that triptolide might be an effective preventive as well as therapeutic agent against prostate cancer and that triptolide may activate a functional p53 pathway in prostate cells.

    View details for Web of Science ID 000177520700029

    View details for PubMedID 12171899

  • Silencing of pi-class glutathione S-transferase in MDA PCa 2a and MDA PCa 2b cells PROSTATE Vidanes, G. M., Paton, V., Wallen, E., Peehl, D. M., Navone, N., Brooks, J. D. 2002; 51 (4): 225-230

    Abstract

    Loss of expression of the glutathione S-transferase-pi (GSTP1) is the most common genetic alteration described in human prostate cancer, occurring in virtually all tumors regardless of grade or stage. Of the available human prostate cancer cell lines, only LNCaP mirrors this phenotype. We investigated whether the prostate cancer cell lines MDA PCa 2a and MDA PCa 2b share this phenotype.GSTP1 protein and mRNA levels were assessed in the MDA PCa 2a and MDA PCa 2b cell lines by Western and Northern blot. DNA methylation was evaluated by Southern blot analysis of genomic DNA digested with the methylation-sensitive restriction enzymes BssHII, NotI, and SacII. Re-expression of GSTP1 was determined by RT-PCR following treatment with 5-azacytidine, a DNA methyltransferase inhibitor, and/or the histone deacetylase inhibitor trichostatin A (TSA).Like all human prostatic carcinomas in vivo, both the MDA PCa 2a and 2b cell lines lack protein and mRNA expression of GSTP1. This lack of expression is associated with methylation in the GSTP1 gene promoter. Treatment with the methyltransferase inhibitor 5-azacytidine resulted in re-expression of GSTP1. By itself, TSA did not result in re-expression of GSTP1, nor did it augment expression induced by 5-azacytidine.MDA PCa 2a and 2b appear to be useful models of human prostatic carcinoma in that they lack expression of GSTP1 due to gene silencing via promoter methylation. Inhibition of histone acetylation does not appear to affect GSTP1 expression.

    View details for DOI 10.1002/pros.10093

    View details for Web of Science ID 000175909900001

    View details for PubMedID 11987150

  • A glucocorticoid-responsive mutant androgen receptor exhibits unique ligand specificity: Therapeutic implications for androgen-independent prostate cancer ENDOCRINOLOGY Krishnan, A. V., Zhao, X. Y., Swami, S., Brive, L., Peehl, D. M., Ely, K. R., Feldman, D. 2002; 143 (5): 1889-1900

    Abstract

    The cortisol/cortisone-responsive AR (AR(ccr)) has two mutations (L701H and T877A) that were found in the MDA PCa human prostate cancer cell lines established from a castrated patient whose metastatic tumor exhibited androgen-independent growth. Cortisol and cortisone bind to the AR(ccr) with high affinity. In the present study, we characterized the structural determinants for ligand binding to the AR(ccr). Our data revealed that many of the C17, C19, and C21 circulating steroids, at concentrations that are found in vivo, functioned as effective activators of the AR(ccr) but had little or no activity via the wild-type AR or GRalpha. Among the synthetic glucocorticoids tested, dexamethasone activated both GRalpha and AR(ccr), whereas triamcinolone was selective for GRalpha. In MDA PCa 2b cells, growth and prostate-specific antigen production were stimulated by potent AR(ccr) agonists such as cortisol or 9alpha-fluorocortisol but not by triamcinolone (which did not bind to or activate the AR(ccr)). Of the potential antagonists tested, bicalutamide (casodex) and GR antagonist RU38486 showed inhibitory activity. We postulate that corticosteroids provide a growth advantage to prostate cancer cells harboring the promiscuous AR(ccr) in androgen-ablated patients and contribute to their transition to androgen-independence. We predict that triamcinolone, a commonly prescribed glucocorticoid, would be a successful therapeutic agent for men with this form of cancer, perhaps in conjunction with the antagonist casodex. We hypothesize that triamcinolone administration would inhibit the hypothalamic-pituitary-adrenal axis, thus suppressing endogenous corticosteroids, which stimulate tumor growth. Triamcinolone, by itself, would not activate the AR(ccr) or promote tumor growth but would provide glucocorticoid activity essential for survival.

    View details for Web of Science ID 000175170500037

    View details for PubMedID 11956172

  • Expression of matrix metalloproteinase-2 and-9 and their inhibitors, tissue inhibitor of metalloproteinase-1 and-2, in primary cultures of human prostatic stromal and epithelial cells JOURNAL OF CELLULAR PHYSIOLOGY Wilson, M. J., Sellers, R. G., Wiehr, C., Melamud, O., Pei, D. Q., Peehl, D. M. 2002; 191 (2): 208-216

    Abstract

    The production of matrix metalloproteinases (MMPs) by prostatic epithelial and/or neighboring stromal cells is considered to be a property that gives cells the capability to penetrate extracellular matrix barriers in normal or neoplastic growth. In order to examine the role of MMPs in the prostate, we evaluated the expression of MMP-2 and -9 and the tissue inhibitors of matrix metalloproteinases (TIMP)-1 and -2 in primary cultures of prostatic stromal and epithelial cells. These cells were isolated from normal tissues of the different zones of the prostate, from benign prostatic hyperplasia (BPH) and from cancer. Stromal cells, regardless of tissue of origin, secreted the 72-kDa proenzyme form of MMP-2, whereas conditioned media (CM) from epithelial cells demonstrated little/no pro-MMP-2 as examined by zymography. Either type of cell did not secrete MMP-9. RT-PCR evaluation showed stromal cells expressed transcripts for MMP-2, but not for MMP-9. Transcripts for MMP-9 were detected in epithelial cells, although no MMP-9 activity was detected in their CM. Treatment of stromal cells with 1 or 10 ng/ml of transforming growth factor-beta (TGF-beta) moderately increased secretion of pro-MMP-2 protein with little change in MMP-2 RNA. However, treatment of epithelial cells with TGF-beta induced expression and secretion of both MMP-2 and-9. The effect of TGF-beta on expression of MMPs by epithelial cells was not duplicated or affected by treatment with insulin-like growth factor (IGF)-1, epidermal growth factor (EGF), platelet-derived growth factor (PDGF), or basic fibroblast growth factor (bFGF). Stromal cells expressed transcripts of both TIMP-1 and -2. Epithelial cells expressed TIMP-1, but little TIMP-2. TGF-beta did not regulate the expression of TIMP-1 or -2 in either stromal or epithelial cells. Our results suggest that the elevated levels of MMP-2 and -9 observed in prostate development and cancer may be due to the elevated TGF-beta associated with these tissues.

    View details for DOI 10.1002/jcp.10092

    View details for Web of Science ID 000174813600010

    View details for PubMedID 12064464

  • Retinol metabolism and lecithin: Retinol acyltransferase levels are reduced in cultured human prostate cancer cells and tissue specimens CANCER RESEARCH Guo, X. J., Knudsen, B. S., Peehl, D. M., Ruiz, A., BOK, D., Rando, R. R., Rhim, J. S., Nanus, D. M., Gudas, L. J. 2002; 62 (6): 1654-1661

    Abstract

    Recent studies from our laboratory have indicated that the metabolism of vitamin A (retinol) to retinyl esters, carried out primarily by the enzyme lecithin:retinol acyltransferase (LRAT), is greatly reduced in human carcinoma cell lines of the oral cavity, skin, breast, and kidney as compared with their normal epithelial counterparts. These studies suggest that human carcinoma cells are retinoid-deficient relative to normal epithelial cells. In this study, we examined the metabolism of [(3)H]retinol and [(3)H]retinoic acid (RA) in human prostate cancer lines and in primary cultures of human prostate epithelial cells. Normal cells esterified all of the [(3)H]retinol added to the cultures. In contrast, all seven prostate cancer cell lines and four primary cultures derived from prostatic adenocarcinomas metabolized only trace amounts of [(3)H]retinol to [(3)H]retinyl esters. Correlated with this relative lack of esterification of [(3)H]retinol by the cancer cells was loss of expression of LRAT protein, whereas normal cells expressed abundant levels of LRAT protein by Western analysis. The metabolism of [(3)H]RA was also examined in these prostatic cells. Two of the prostate cancer tumor lines, DU 145 and PJ-1, exhibited rapid metabolism of [(3)H]RA; in contrast, the other tumor lines or primary cultures metabolized [(3)H]RA at a much slower rate. We also found that the immortalization of normal human prostatic epithelial cells by SV40 T antigen led to a reduction in LRAT protein expression and esterification of [(3)H]retinol. Further transformation to tumorigenicity with the ras oncogene resulted in loss of detectable LRAT expression. Finally, we analyzed LRAT protein expression in tissue sections from six prostatectomy specimens by immunohistochemistry. LRAT protein was predominantly expressed in the basal cells of normal prostatic epithelium, whereas its expression was lost in prostate cancer. Collectively, these data implicate aberrant retinoid metabolism in the process of prostatic carcinogenesis.

    View details for Web of Science ID 000174560200013

    View details for PubMedID 11912136

  • Role of glutathione depletion and reactive oxygen species generation in apoptotic signaling in a human B lymphoma cell line CELL DEATH AND DIFFERENTIATION Armstrong, J. S., Steinauer, K. K., Hornung, B., Irish, J. M., Lecane, P., Birrell, G. W., Peehl, D. M., Knox, S. J. 2002; 9 (3): 252-263

    Abstract

    The primary objective of this study was to determine the sequence of biochemical signaling events that occur after modulation of the cellular redox state in the B cell lymphoma line, PW, with emphasis on the role of mitochondrial signaling. L-Buthionine sulphoximine (BSO), which inhibits gamma glutamyl cysteine synthetase (gammaGCS), was used to modulate the cellular redox status. The sequence and role of mitochondrial events and downstream apoptotic signals and mediators was studied. After BSO treatment, there was an early decline in cellular glutathione (GSH), followed by an increase in reactive oxygen species (ROS) production, which induced a variety of apoptotic signals (detectable at different time points) in the absence of any external apoptotic stimuli. The sequence of biochemical events accompanying apoptosis included a 95% decrease in total GSH and a partial (25%) preservation of mitochondrial GSH, without a significant increase in ROS production at 24h. Early activation and nuclear translocation of the nuclear factor kappa B subunit Rel A was observed at approximately 3h after BSO treatment. Cytochrome c release into the cytosol was also seen after 24h of BSO treatment. p53 protein expression was unchanged after redox modulation for up to 72 h, and p21waf1 independent loss of cellular proliferation was observed. Surprisingly, a truncated form of p53 was expressed in a time-dependent manner, beginning at 24h after BSO incubation. Irreversible commitment to apoptosis occurred between 48 and 72 h after BSO treatment when mitochondrial GSH was depleted, and there was an increase in ROS production. Procaspase 3 protein levels showed a time-dependent reduction following incubation with BSO, notably after 48 h, that corresponded with increasing ROS levels. At 96 h, caspase 3 cleavage products were detectable. The pan-caspase inhibitor zVADfmk, partially blocked the induction of apoptosis at 48 h, and was ineffective after 72 h. PW cells could be rescued from apoptosis by removing them from BSO after up to 48, but not 72 h incubation with BSO. Mitochondrial transmembrane potential (DeltaPsi(m)) remained intact in most of the cells during the 72 h observation period, indicating that DeltaPsi(m) dissipation is not an early signal for the induction of redox dependent apoptosis in PW cells. These data suggest that a decrease in GSH alone can act as a potent early activator of apoptotic signaling. Increased ROS production following mitochondrial GSH depletion, represents a crucial event, which irreversibly commits PW cells to apoptosis.

    View details for DOI 10.1038/sj.cdd.4400959

    View details for Web of Science ID 000174032600006

    View details for PubMedID 11859408

  • Cadmium-induced neoplastic transformation of human prostate epithelial cells INTERNATIONAL JOURNAL OF ONCOLOGY Nakamura, K., Yasunaga, Y., Ko, D., Xu, L. L., Moul, J. W., Peehl, D. M., Srivastava, S., Rhim, J. S. 2002; 20 (3): 543-547

    Abstract

    Cadmium is a ubiquitous environmental human carcinogen. Epidemiological and animal studies have suggested its carcinogenic potential on the prostate. In the present study, non-tumorigenic human prostate epithelial cells (pRNS-1-1) immortalized by simian papovavirus (SV40) were transformed after repeated exposures to cadmium. Such transformants showed morphological alterations, anchorage-independent growth in soft agar, and formed tumors when transplanted into SCID mice. The tumors were characterized histologically as poorly-differentiated adenocarcinomas, expressing prostate-specific antigen (PSA), androgen receptor (AR), prostate stem cell antigen (PSCA), NKX3.1 and cytokeratin 8 (CK8). These findings provide evidence of malignant transformation of human prostate epithelial cells exposed to this environmentally important chemical.

    View details for Web of Science ID 000173883400014

    View details for PubMedID 11836566

  • Expression of peroxisome proliferator activated receptor gamma (PPAR gamma) regulated secretary genes in benign and malignant prostate tissues Iyengar, S., Roberts, R. L., Manning, S., Xu, Y., Shook, M. F., Olson, S. J., Wheeler, T. M., Peehl, D. L., Shappell, S. B. NATURE PUBLISHING GROUP. 2002: 165A–166A
  • Expression of peroxisome proliferator activated receptor gamma (PPAR gamma) regulated secretory genes in benign and malignant prostate tissues Iyengar, S., Roberts, R. L., Manning, S., Xu, Y., Shook, M. F., Olson, S. J., Wheeler, T. M., Peehl, D. L., Shappell, S. B. NATURE PUBLISHING GROUP. 2002: 165A–166A
  • Genetic polymorphisms in the androgen receptor and type II 5 alpha-reductase genes in prostate enlargement JOURNAL OF UROLOGY Shibata, A., Stamey, T. A., McNeal, J. E., Cheng, I., Peehl, D. M. 2001; 166 (4): 1560-1564

    Abstract

    We examined the association of androgen receptor gene cytosine-adenine-guanine (CAG) repeat length and the 2 single nucleotide polymorphisms A49T and V89L in the type II 5 alpha-reductase gene with prostate enlargement measured as the weight of the surgically removed prostate.A total of 68 men with a prostate weighing 80 gm. or greater were compared with 197 controls with a prostate weighing less than 80 gm. These men had undergone radical prostatectomy between 1992 and 1996. DNA was extracted from archival prostate tissue uninvolved with cancer and genotyped for 3 polymorphic markers. The effects of genetic variants and clinicopathological variables on prostate enlargement risk were estimated by logistic regression.The age adjusted odds ratio estimate of prostate enlargement risk in men with 23 or greater versus 20 or fewer CAG repeats was 0.41 (95% confidence interval 0.19 to 0.89). This risk reduction was consistently found when an alternative prostate enlargement definition and subject restriction were used. No consistent association with prostate enlargement risk was observed for A49T or V89L polymorphisms.Our finding further supports the hypothesis that the shorter CAG repeat length of the androgen receptor gene is related to prostate enlargement.

    View details for Web of Science ID 000170950100102

    View details for PubMedID 11547131

  • Prostatic expression of hensin, a protein implicated in epithelial terminal differentiation PROSTATE Ma, J. F., Takito, J., Vijayakumar, S., Peehl, D. M., Olsson, C. A., Al-Awqati, Q. 2001; 49 (1): 9-18

    Abstract

    Hensin induces terminal differentiation in rabbit kidney collecting tubule cells. Rabbit hensin and human DMBT1 result from alternative splicing of the same gene. The human DMBT1 gene is located on chromosome 10q25-26, a region often deleted in prostate cancer. In this study we examined the potential role of this gene in terminal differentiation of prostate, as well as its role in prostatic carcinogenesis.We searched for deletions of this gene in prostatic cells cultured from cancer and benign tissues using PCR and cDNA cloning. The expression of hensin/DMBT1 in cultured cells and during prostate development was characterized by immunochemistry.No deletions of hensin/DMBT1 similar to those found in glioblastomas, lung and esophageal cancers were observed in prostate cancer or BPH cells. Hensin/DMBT1 protein was localized in intracellular vesicles of epithelial cells in neonatal and 6-week-old mouse prostates. By 6 weeks, hensin/DMBT1 began to localize in the basal lamina of the prostate and vas deferens. In matured 6-month-old prostates, there was extensive deposition of hensin/DMBT1 in the basal lamina.There is no evidence that hensin/DMBT1 is implicated in prostatic carcinogenesis. The localization of hensin/DMBT1 during maturation raises the possibility that hensin/DMBT1 is involved in terminal differentiation of the prostate and vas deferens.

    View details for Web of Science ID 000170784100002

    View details for PubMedID 11550206

  • Characterization of cultured human prostatic epithelial cells by cluster designation antigen expression CELL AND TISSUE RESEARCH Liu, A. Y., Peehl, D. M. 2001; 305 (3): 389-397

    Abstract

    Cultured prostatic epithelial cells have been extensively studied as a model of prostate biology. What is the lineage relationship of the cultured cells to the epithelial cell types in tissue? How different are cultured cells derived from tumor tissue to those derived from benign tissue? Expression of cluster designation (CD) cell surface molecules has been shown to be useful in characterizing cells according to lineage. A CD profile was therefore generated for cultured human prostatic epithelial cells and compared with those previously established for basal and luminal epithelial cells in the prostate. Presence of CD44, CD49b, CD49f, and CD104 and absence of CD57 suggests that cultured cells were derived from basal cells of prostatic tissues. However, expression of certain CD antigens characteristic of luminal epithelial cells was also observed in subpopulations of cultured cells. The pattern of CD antigens in cultured cells reflects a phenotype similar to that of transit-amplifying cells that have been described in the prostate. Several CD antigens were found expressed by both cultured prostatic epithelial and stromal cells, and are probably associated with cell proliferation. The CD profiles of cultured epithelial cell strains derived from normal compared with malignant tissues were notably similar to each other and to that of the prostate cancer cell line PC-3. We conclude that cells in culture retain expression of certain lineage-characteristic CD antigens. Furthermore, CD antigens can define subpopulations of cells with differential gene expression.

    View details for Web of Science ID 000170888100011

    View details for PubMedID 11572092

  • Rationale for combination ketoconazole/ vitamin D treatment of prostate cancer. Urology Peehl, D. M., Seto, E., Feldman, D. 2001; 58 (2): 123-126

    Abstract

    The high rate of progression of prostate cancer after androgen deprivation therapy mandates that new strategies be developed. Adjuvant therapy combined with androgen deprivation may slow or prevent progression. Ketoconazole plus calcitriol therapy is an example of 1 such a combination with a mechanistic basis for synergistic activity. Ketoconazole is commonly used as a second-line androgen deprivation therapy. This imidazole derivative is an inhibitor of P-450 enzymes, including those involved in steroidogenesis. Other P-450 enzymes that are inhibited by ketoconazole include 1alpha-hydroxylase and 24-hydroxylase, which metabolize vitamin D. Growth inhibition of prostate cancer cells by vitamin D depends on levels of the active metabolite, 1,25-dihydroxyvitamin D(3) (calcitriol). The enzyme 24-hydroxylase converts calcitriol to less active products. The inhibition of 24-hydroxylase by ketoconazole maintains the magnitude and duration of response to calcitriol. Combined ketoconazole/calcitriol therapy might therefore potentiate the antitumor activity of calcitriol. Because androgen-independent prostate cancer cells often remain responsive to growth inhibition by calcitriol, it is also possible that calcitriol would slow or prevent development of androgen-independent cancer growth. Another consideration is that ketoconazole blocks 1alpha-hydroxylase activity, which is the key enzyme that creates calcitriol in the body. Therefore, patients receiving ketoconazole therapy are likely to be deficient in vitamin D. The detrimental consequences of vitamin D deficiency in these patients would also be alleviated by the addition of calcitriol to the therapeutic regimen.

    View details for PubMedID 11502466

  • Rationale for combination ketoconazole/vitamin D treatment of prostate cancer 1st International Conference on Newer Approaches to Androgen Deprivation Therapy in Prostate Cancer Peehl, D. M., Seto, E., Feldman, D. ELSEVIER SCIENCE INC. 2001: 123–26

    Abstract

    The high rate of progression of prostate cancer after androgen deprivation therapy mandates that new strategies be developed. Adjuvant therapy combined with androgen deprivation may slow or prevent progression. Ketoconazole plus calcitriol therapy is an example of 1 such a combination with a mechanistic basis for synergistic activity. Ketoconazole is commonly used as a second-line androgen deprivation therapy. This imidazole derivative is an inhibitor of P-450 enzymes, including those involved in steroidogenesis. Other P-450 enzymes that are inhibited by ketoconazole include 1alpha-hydroxylase and 24-hydroxylase, which metabolize vitamin D. Growth inhibition of prostate cancer cells by vitamin D depends on levels of the active metabolite, 1,25-dihydroxyvitamin D(3) (calcitriol). The enzyme 24-hydroxylase converts calcitriol to less active products. The inhibition of 24-hydroxylase by ketoconazole maintains the magnitude and duration of response to calcitriol. Combined ketoconazole/calcitriol therapy might therefore potentiate the antitumor activity of calcitriol. Because androgen-independent prostate cancer cells often remain responsive to growth inhibition by calcitriol, it is also possible that calcitriol would slow or prevent development of androgen-independent cancer growth. Another consideration is that ketoconazole blocks 1alpha-hydroxylase activity, which is the key enzyme that creates calcitriol in the body. Therefore, patients receiving ketoconazole therapy are likely to be deficient in vitamin D. The detrimental consequences of vitamin D deficiency in these patients would also be alleviated by the addition of calcitriol to the therapeutic regimen.

    View details for Web of Science ID 000170422300033

  • Activation of EphA receptor tyrosine kinase inhibits the Ras/MAPK pathway NATURE CELL BIOLOGY Miao, H., Wei, B. R., Peehl, D. M., Li, Q., Alexandrou, T., Schelling, J. R., Rhim, J. S., Sedor, J. R., Burnett, E., Wang, B. C. 2001; 3 (5): 527-530

    Abstract

    Interactions between Eph receptor tyrosine kinases (RTKs) and membrane-anchored ephrin ligands critically regulate axon pathfinding and development of the cardiovascular system, as well as migration of neural cells. Similar to other RTKs, ligand-activated Eph kinases recruit multiple signalling and adaptor proteins, several of which are involved in growth regulation. However, in contrast to other RTKs, activation of Eph receptors fails to promote cell proliferation or to transform rodent fibroblasts, indicating that Eph kinases may initiate signalling pathways that are distinct from those transmitted by other RTKs. Here we show that stimulation of endogenous EphA kinases with ephrin-A1 potently inhibits the Ras/MAPK cascade in a range of cell types, and attenuates activation of mitogen-activated protein kinase (MAPK) by receptors for platelet-derived growth factor (PDGF), epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF). In prostatic epithelial cells and endothelial cells, but not fibroblasts, treatment with ephrin-A1 inhibits cell proliferation. Our results identify EphA kinases as negative regulators of the Ras/MAPK pathway that exert anti-mitogenic functions in a cell-type-specific manner.

    View details for Web of Science ID 000168592500026

    View details for PubMedID 11331884

  • Reduced 1 alpha-hydroxylase activity in human prostate cancer cells correlates with decreased susceptibility to 25-hydroxyvitamin D-3-induced growth inhibition CANCER RESEARCH Hsu, J. Y., Feldman, D., McNeal, J. E., Peehl, D. M. 2001; 61 (7): 2852-2856

    Abstract

    Evidence from epidemiological, molecular, and genetic studies suggests a role for vitamin D in the development and/or progression of prostate cancer. In experimental models and clinical trials, 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] was shown to exert antiproliferative, prodifferentiating, and antimetastatic/invasive effects on prostatic epithelial cells. Because the direct clinical application of 1,25(OH)2D3 is limited by the major side effect of hypercalcemia, we investigated the potential therapeutic utility of its less calcemic precursor, 25-hydroxyvitamin D3 [25(OH)D3], which is converted locally within the prostate to 1,25(OH)2D3 by 1alpha-hydroxylase. Quantification of 1alpha-hydroxylase activity in human prostatic epithelial cells by enzyme-substrate reaction analyses revealed a significantly decreased activity in cells derived from adenocarcinomas compared with cells derived from normal tissues or benign prostatic hyperplasia (BPH). In growth assays, we found that 25(OH)D3 inhibited growth of normal or BPH cells similarly to 1,25(OH)2D3. In contrast, in primary cultures of cancer cells and established cell lines, the antiproliferative action of 25(OH)D3 was significantly less pronounced than that of 1,25(OH)2D3. Our results indicate that growth inhibition by 25(OH)D3 depends on endogenous 1alpha-hydroxylase activity, and that this activity is deficient in prostate cancer cells. This finding has ramifications for both the prevention and therapy of prostate cancer with vitamin D compounds.

    View details for Web of Science ID 000167967100012

    View details for PubMedID 11306457

  • SMAD3 represses androgen receptor-mediated transcription CANCER RESEARCH Hayes, S. A., Zarnegar, M., Sharma, M., Yang, F. J., Peehl, D. M., ten Dijke, P., Sun, Z. J. 2001; 61 (5): 2112-2118

    Abstract

    The androgen-signaling pathway is important in the growth and progression of prostate cancer. Androgen ablation therapy, which may result in programmed cell death, is often used to treat advanced prostate cancer. The growth-promoting effects of androgen are mediated mostly through the androgen receptor (AR). Transforming growth factor beta (TGF-beta) plays critical roles in controlling prostate cell proliferation, differentiation, and apoptosis. Normal transcripts and proteins of TGF-beta receptors are frequently lost in prostate cancer cells, especially in advanced stages of the disease. However, the mechanisms by which TGF-beta inhibits proliferation and induces apoptosis in prostate cancer cells is not clear. We investigated the molecular mechanism by which TGF-beta inhibits transcriptional activation mediated by AR. Using transient transfection systems, we demonstrated that Smad3 specifically represses transcriptional activation mediated by AR on two natural androgen-responsive promoters. This repression is transmitted through TGF-beta signaling and can be regulated by other Smad proteins. A protein-protein interaction between AR and Smad3 was identified in vitro and in vivo, and the transcription activation domain of AR and the MH2 of Smad3 were identified as being responsible for binding. Additional functional experiments showed that the repression of AR by Smad3 is mediated solely through the MH2 domain. These results provide fresh insight for understanding the mechanism by which TGF-beta regulates the androgen-signaling pathway in prostate cancer cells.

    View details for Web of Science ID 000167568100056

    View details for PubMedID 11280774

  • Cultured stromal cells: An in vitro model of prostatic mesenchymal biology PROSTATE Peehl, D. M., Sellers, R. G. 2000; 45 (2): 115-123

    Abstract

    Initial efforts to develop in vitro models to study prostatic biology focused on the culture and characterization of epithelial cells. Recently, attention has turned towards inclusion of stromal cells in experimental systems.Improved methods to isolate and culture stromal cells have been developed. An array of markers are employed to characterize subtypes of stromal cells, with particular interest in smooth muscle differentiation.Defined, serum-free media are available for certain experimental applications. Conditions that promote smooth muscle differentiation have been identified. Investigators have characterized hormonal and peptide factors that regulate the growth of prostatic stromal cells, and have also described paracrine factors produced by stromal cells that influence epithelial biology.Prostatic stromal-cell cultures are now widely employed by a large number of investigators for a diverse array of experimental purposes. While further refinement is required to obtain model systems that fully mimic in vivo processes, the availability of stromal- and epithelial-cell cultures provides a valuable resource for studying normal prostatic biology as well as benign prostatic hyperplasia (BPH) and cancer.

    View details for Web of Science ID 000089614600005

    View details for PubMedID 11027410

  • Brefeldin A induces p53-independent apoptosis in primary cultures of human prostatic cancer cells JOURNAL OF UROLOGY Wallen, E., Sellers, R. G., Peehl, D. M. 2000; 164 (3): 836-841

    Abstract

    The objective of this study was to investigate growth-inhibitory and apoptotic activity of the experimental antitumor drug, brefeldin A (BFA), on primary cultures of human epithelial cells derived from prostatic adenocarcinomas.Clonal assays were performed to evaluate the effects of BFA on growth of prostatic cancer cell strains. Loss of cell viability in response to BFA was assessed by trypan blue exclusion. Induction of apoptosis by BFA was evaluated by morphologic criteria, electrophoretic assay of DNA fragmentation, and a cell death ELISA. Immunoblots were used to monitor p53 and pRB expression in response to BFA.BFA was growth-inhibitory at a half-maximal concentration of 5 ng./ml. (18 nM). Morphological manifestations of apoptosis were evident by 24 hours of treatment. Cell viability declined and the cell death ELISA indicated an 18-fold increase in apoptosis in BFA-treated versus untreated cells at 48 hours. DNA fragmentation was also seen at 48 hours. Levels of p53 were not altered by BFA, but pRB was maintained in a hypophosphorylated state by BFA treatment.BFA is a potent inducer of apoptosis in prostatic cancer cells via a p53-independent mechanism. Cells derived from low-grade as well as high-grade cancers responded similarly to BFA. Since p53-mediated pathways of apoptosis may frequently be abrogated in prostatic cancer cells, agents such as BFA that induce p53-independent cell death may be promising candidates for chemotherapeutic agents.

    View details for Web of Science ID 000088682100068

    View details for PubMedID 10953164

  • Stepwise genetic changes associated with progression of nontumorigenic HPV-18 immortalized human prostate cancer-derived cell line to a malignant phenotype CANCER GENETICS AND CYTOGENETICS Hukku, B., Mally, M., Cher, M. L., Peehl, D. M., Kung, H. F., Rhim, J. S. 2000; 120 (2): 117-126

    Abstract

    Cytogenetics, fluorescence in situ hybridization (FISH), and comparative genomic hybridization (CGH) were used to identify genes that are involved in the development and progression of prostate cancer. For that purpose, we chose a cell line established in vitro from a prostatic adenocarcinoma which was nontumorigenic in nude mice and followed its progression to a tumorigenic cell line. Stepwise changes were observed in the cell line as it became tumorigenic. The composite karyotype at the nontumorigenic stage (CA-HPV-10) was 68 approximately 77,XXY,-(1, 9, 13, 14, 19, 22),+(4, 5, 11, 18, 20, 21),+(del(1) (q23q31)=M1 (two copies), +der(9)t(1;9)(q24 approximately q31;p23)=M5(two copies), der(14)t(14;?)(q10;?)=M17 in the majority of metaphases. These two derivative chromosomes were also observed a previous study. Our CGH analysis clearly showed that this deleted region in M1 is, in fact, translocated with derivative M5 and, in reality, is amplified. The cell line established from nodule (SCID 5019 p11), showed a number of new changes, as described; however, the most significant change was amplification of the 8q23 approximately qter region, harboring c-myc. This region was translocated with chromosomes 2, 4, and 16 as der(2)t(2;8)(q33;q23)=M12, der(4)t(4;8)(q34;q23)=M11, and der(16)t(8;16)(q24;q21)=M9. We deduce from our study that amplification of c-myc and other genes in the 8q23 approximately qter region were important in progression but did not lead to tumorigenicity. The population that became tumorigenic (SCID 5019 II) showed almost all of the same changes in the karyotype as observed in the nodular cell line; the only significant change was the appearance of der(11)t(4;11)(q32;q22)=M7 and the addition of another copy of t(3q;7p)=M2. These new changes lead to loss of chromosomes 3p, 4pter approximately q34, 6, 7q21 approximately qter, 11q22 approximately qter, and 18q, and gain of 3q, 7p, 8q23 approximately qter, and 11pter approximately q22, before the cell line became tumorigenic. The clonal selection of the population is proven by the presence of a number of the same derivative chromosomes in both the nodular and tumorigenic cell line. As it progressed to tumorigenicity, some of the same changes observed in the original study re-appear at different stages of malignancy, although it was absent in the nontumorigenic cell line. These are: der(16)t(8;16)(q24;q21)=M9 in the nodular cell line and der(11)t(4;11)(q32;q22)=M7 in the tumorigenic cell line. In our system, amplification of c-myc and other genes in der(2)t(2;8)(q33;q23)=M12,der(4) t(4;8)(q34;q23)=M11 together with the presence of der(16)t(8;16)(q24;q21)=M9 and der(11)t(4;11)(q32;q22)=M5 makes the cell line tumorigenic. It is either nontumorigenic, with the presence of a marker equivalent to der(16)=M9 and der(11)=M7 observed in the original study, and only nodular (SCID 5019 p11, present study), with the presence of number of markers with c-myc amplification (M9, M11, and M12). There is accumulation of all the above-mentioned changes in the same cell before it becomes tumorigenic.

    View details for Web of Science ID 000088925000005

    View details for PubMedID 10942801

  • 1 alpha,25-dihydroxyvitamin D-3 inhibits prostate cancer cell growth by androgen-dependent and androgen-independent mechanisms ENDOCRINOLOGY Zhao, X. Y., Peehl, D. M., Navone, N. M., Feldman, D. 2000; 141 (7): 2548-2556

    Abstract

    We recently reported that 1alpha,25-dihydroxyvitamin D3 [1,25-(OH)2D3] inhibits the growth of the LNCaP human prostate cancer cell line by an androgen-dependent mechanism. In the present study we examined the actions and interactions of 1,25-(OH)2D3 and the androgen 5alpha-dihydrotestosterone (DHT) on two new human prostate cancer cell lines (MDA), MDA PCa 2a and MDA PCa 2b. Scatchard analyses revealed that both cell lines express high affinity vitamin D receptors (VDRs) with a binding affinity (Kd) for [3H]1,25-(OH)2D3 of 0.1 nM. However, the MDA cell lines contain low affinity androgen receptors (ARs) with a Kd of 25 nM for [3H]DHT binding. This is 50-fold lower than the AR in LNCaP cells (Kd = 0.5 nM). Their response to DHT is greatly reduced; 2a cells do not respond to 100 nM DHT, and 2b cells show a modest response at that high concentration. 1,25-(OH)2D3 causes significant growth inhibition in both MDA cell lines, greater (for 2b cells) or lesser (for 2a cells) than that in the LNCaP cell line. Moreover, 1,25-(OH)2D3 significantly up-regulates AR messenger RNA in all three cell lines, as shown by Northern blot analysis. The growth inhibitory effect of 1,25-(OH)2D3 on LNCaP cells is blocked by the pure antiandrogen, Casodex, as we previously reported. However, Casodex (at 1 microM) did not block the antiproliferative activity of 1,25-(OH)2D3 in MDA cells. In conclusion, the growth inhibitory action of 1,25-(OH)2D3 in the MDA cell lines appears to be androgen independent, whereas the actions of 1,25-(OH)2D3 in LNCaP cells are androgen dependent. Most importantly, the MDA cell lines, derived from a bone metastasis of human prostate carcinoma, remain sensitive to 1,25-(OH)2D3, a finding relevant to the therapeutic application of vitamin D and its low calcemic analogs in the treatment of advanced prostate cancer.

    View details for Web of Science ID 000088386200032

    View details for PubMedID 10875257

  • Expression of senescence-associate beta-galactosidase in enlarged prostates from men with benign prostatic hyperplasia UROLOGY Choi, J., Shendrik, Peacocke, M., Peehl, D., Buttyan, R., Ikeguchi, E. F., Katz, A. E., Benson, M. C. 2000; 56 (1): 160–66

    Abstract

    Cellular senescence is a unique cellular response pathway thought to be closely associated with the aging process. The senescent phenotype is characterized by the loss of a cell's ability to respond to proliferative and apoptotic stimuli even while normal metabolic activity and vitality is maintained. Recently, a novel biomarker, senescent-associated beta-galactosidase (SA-beta-gal), was found to identify cells with the senescent phenotype. In the present study, we examined whether human prostatic epithelial cells adopt a senescence-associated phenotype after prolonged culture and analyzed a series of human benign prostatic hyperplasia (BPH) specimens to determine whether the cellular senescence process might be a factor in the development of BPH.A primary culture of epithelial cells was established from the normal tissue of the peripheral zone of a radical prostatectomy specimen and was serially passaged until senescence. Forty-three human prostate specimens were obtained subsequent to radical prostatectomy or transrectal ultrasound-guided biopsy. The cultured cells and tissue specimens were histochemically stained to reveal the expression of SA-beta-gal, the cellular senescence biomarker.As has been reported for other types of cultured cells, human prostatic epithelial cells demonstrated widespread expression of the cellular senescence marker, SA-beta-gal, on prolonged culture. In our survey of hypertrophied human prostate tissues, 17 specimens (40%) of the 43 analyzed demonstrated positive staining for SA-beta-gal. In these tissues, SA-beta-gal expression was noted only in the epithelial cells. No statistical correlation (P = 0.42) between the chronologic age of the patient donor and SA-beta-gal expression was found. However, a high prostate weight (greater than 55 g) was found to correlate strongly with the expression of the SA-beta-gal biomarker (P = 0. 0001).Cultured prostatic epithelial cells expressed SA-beta-gal on reaching replicative senescence in vitro. The survey of human BPH specimens for the senescent marker showed that prostatic epithelial cells in patients with BPH with more advanced enlargement of the prostate (greater than 55 g prostate weight) expressed SA-beta-gal, and the prostates from patients with BPH that weighed less than 55 g tended to lack senescent epithelial cells. On the basis of these results, we propose that the accumulation of senescent epithelial cells may play a role in the development of the prostatic enlargement associated with BPH.

    View details for DOI 10.1016/S0090-4295(00)00538-0

    View details for Web of Science ID 000087900600044

    View details for PubMedID 10869659

  • Glucocorticoids can promote androgen-independent growth of prostate cancer cells through a mutated androgen receptor NATURE MEDICINE Zhao, X. Y., Malloy, P. J., Krishnan, A. V., Swami, S., Navone, N. M., Peehl, D. M., Feldman, D. 2000; 6 (6): 703-706

    Abstract

    The androgen receptor (AR) is involved in the development, growth and progression of prostate cancer (CaP). CaP often progresses from an androgen-dependent to an androgen-independent tumor, making androgen ablation therapy ineffective. However, the mechanisms for the development of androgen-independent CaP are unclear. More than 80% of clinically androgen-independent prostate tumors show high levels of AR expression. In some CaPs, AR levels are increased because of gene amplification and/or overexpression, whereas in others, the AR is mutated. Nonetheless, the involvement of the AR in the transition of CaP to androgen-independent growth and the subsequent failure of endocrine therapy are not fully understood. Here we show that in CaP cells from a patient who failed androgen ablation therapy, a doubly mutated AR functioned as a high-affinity cortisol/cortisone receptor (ARccr). Cortisol, the main circulating glucocorticoid, and its metabolite, cortisone, both equally stimulate the growth of these CaP cells and increase the secretion of prostate-specific antigen in the absence of androgens. The physiological concentrations of free cortisol and total cortisone in men greatly exceed the binding affinity of the ARccr and would activate the receptor, promoting CaP cell proliferation. Our data demonstrate a previously unknown mechanism for the androgen-independent growth of advanced CaP. Understanding this mechanism and recognizing the presence of glucocorticoid-responsive AR mutants are important for the development of new forms of therapy for the treatment of this subset of CaP.

    View details for Web of Science ID 000087438300044

    View details for PubMedID 10835690

  • p16(INK4A) mediates cyclin dependent kinase 4 and 6 inhibition in senescent prostatic epithelial cells CANCER RESEARCH Sandhu, C., Peehl, D. M., Slingerland, J. 2000; 60 (10): 2616-2622

    Abstract

    The senescence checkpoint constrains the proliferative potential of normal cells in culture to a finite number of cell doublings. In this study, we investigated the mechanism of cyclin dependent kinase (cdk) inhibition in senescent human prostatic epithelial cells (HPECs). Progression of HPECs from early passage to senescence was accompanied by a gradual loss of cells in S phase and an accumulation of cells containing 2N DNA. Furthermore, G1-S phase-associated kinase activities progressively diminished with increasing cell passage. In senescent HPECs, cdk4 and cyclin E1- and A-associated kinases were catalytically inactive. In contrast to observations in senescent fibroblasts, levels of the kinase inhibitor protein (KIP) inhibitor p21CIP1 diminished over the proliferative life span of HPECs. p27KIP1 levels fell as cells approached senescence, and the association of both p21CIP1 and p27KIP1 with cdk4/6 complexes was decreased. However, the level of cyclin E1-associated KIP molecules was unaltered as cells progressed into senescence. Progression to senescence was accompanied by a progressive increase in both the level of p16(INK4A) and in its association with cdk4 and cdk6. As HPECs approached senescence, cdk4- and cdk6-bound p16(INK4A) showed a shift to a slower mobility due to a change in its phosphorylation profile. As p16(INK4A) increased in cdk4 and cdk6 complexes, there was a loss of cyclin D1 binding. The altered phosphorylation of p16(INK4A) in senescent prostatic epithelial cells may facilitate its association with cdk4 and cdk6 and play a role in the inactivation of these kinases.

    View details for Web of Science ID 000087059600012

    View details for PubMedID 10825132

  • Transforming growth factor-beta induces growth inhibition and IGF-binding protein-3 production in prostatic stromal cells: abnormalities in cells cultured from benign prostatic hyperplasia tissues JOURNAL OF ENDOCRINOLOGY Cohen, P., Nunn, S. E., Peehl, D. M. 2000; 164 (2): 215-223

    Abstract

    The IGF axis has been implicated in the pathogenesis of benign prostatic hyperplasia (BPH) via the paracrine action of IGFs and IGF-binding proteins (IGFBPs). In this study, we examined the regulation of cell growth and IGFBP-3 secretion by transforming growth factor-beta (TGF-beta) in prostatic stromal cell (PC-S) cultures from histologically normal tissues and tissues from BPH. PC-S cultures were treated with varying doses of TGF-beta1. Forty-eight hour conditioned media (CM) from these cultures were subjected to Western immunoblotting and ligand blotting for detection and quantification of IGFBPs. IGFBPs-2, -3 and -4 were detected in the CM from normal PC-S cultures. In CM from BPH PC-S, IGFBP-3 levels were 2-fold lower at baseline than in the normal PC-S CM, in addition to the differences in IGFBPs-2 and -5 which we have previously reported. In response to TGF-beta1, a 15-fold increase in the levels of IGFBP-3 was observed in normal PC-S CM, while a mere 2-fold increase was observed in BPH PC-S CM (P<0.001). These findings were confirmed by specific immunoblotting and immunocytochemistry. IGFBP-3 mRNA levels detected by Northern blotting of total RNA extracted from similar cultures showed the induction of IGFBP-3 expression by TGF-beta1 in normal PC-S and its lack of induction in BPH PC-S. Cell growth inhibition in response to TGF-beta1 correlated with the IGFBP-3 concentrations found in CM. Normal PC-S showed a 60% decrease in cell number after 10 days in media with 1 ng/ml TGF-beta1, compared with the untreated control. The decrease in proliferation observed in comparably treated BPH cells was only 20% (P<0.001). In conclusion, BPH PC-S had a reduced IGFBP-3 response to TGF-beta1 and demonstrated decreased TGF-beta1-induced growth inhibition relative to normal PC-S. We hypothesize that in normal PC-S, TGF-beta exerts its anti-proliferative effects by stimulating the production of IGFBP-3, which acts as an inhibitory factor, either by inhibiting IGFs or directly by interacting with cells, and that this process is altered in BPH PC-S.

    View details for Web of Science ID 000085487200012

    View details for PubMedID 10657857

  • Two mutations identified in the androgen receptor of the new human prostate cancer cell line MDA PCa 2a JOURNAL OF UROLOGY Zhao, X. Y., Boyle, B., Krishnan, A. V., Navone, N. M., Peehl, D. M., Feldman, D. 1999; 162 (6): 2192-2199

    Abstract

    We have characterized the androgen receptor (AR) in a new human prostate cancer cell line, MDA PCa 2a, that has recently been established from a bone metastasis of a patient whose cancer exhibited androgen-independent growth.Androgen responsiveness of these cells was assessed by measuring the effect of DHT and R1881 on cell growth and PSA secretion. Scatchard analysis was used to characterize the affinity and abundance of AR protein. Using a PCR based strategy, genomic DNA of the entire coding region of AR gene was sequenced to identify possible mutations.These cells express abundant AR (Nmax = 685 +/- 149 fmol./mg. protein), but the AR binding affinity (Kd) for DHT is only 25 nM, approximately 50-fold lower affinity than the mutated AR in LNCaP prostate cancer cells (Kd = 0.5 nM) or the wildtype AR in MCF-7 breast cancer cells (Kd = 0.4 nM). Two mutations, L701H and T877A, were identified in the ligand binding domain of the AR gene. Compared with LNCaP cells, the new cell line is significantly less responsive to DHT and R1881 as well as to other androgens such as testosterone, androstenedione, and DHEA. Similar to LNCaP cells, the ligand specificity of the AR in MDA PCa 2a cells appears to be relaxed and non-androgens such as progesterone and estradiol act as agonists although with less potency than in LNCaP cells. Interestingly, in the absence of androgens, the new cell line expresses 15-fold higher baseline levels of PSA than LNCaP.Two mutations were identified in the AR gene of the MDA PCa 2a cell line that are likely responsible for the decreased androgen sensitivity and altered ligand specificity observed in these cells. Thus, this new cell line with partial androgen responsiveness and PSA expression can serve as a functionally relevant model system of bone metastatic prostate cancer, and can be used to investigate the role of AR mutations in prostate cancer and its progression to androgen independence.

    View details for Web of Science ID 000083639500119

    View details for PubMedID 10569618

  • Altered growth regulation and loss of response to retinoic acid accompany tumorigenic transformation of prostatic cells ANTICANCER RESEARCH Peehl, D. M., Sellers, R. G., Arnstein, P., Kung, H. F., Rhim, J. S. 1999; 19 (5B): 3857-3864

    Abstract

    In vitro models of human prostatic carcinogenesis are increasingly available and include representatives of normal, immortal, tumorigenic and metastatic phenotypes. In this study, growth regulation of immortal, but non-tumorigenic, human papillomavirus-transformed prostatic epithelial cells was compared to that of their tumorigenic variants. These variants were created either by exposure to a carcinogen or by passage through mice. In all cases, tumorigenic cells retained responsiveness to a potent mitogen, epidermal growth factor, and to a potent growth inhibitory factor, 1,25-dihydroxyvitamin D3. Responses to other growth regulatory factors were altered. One set of transformants, CA-HPV-10 and its tumorigenic variants 5019 and 5019IIc, lost their requirement for insulin-like growth factor. Another set, RWPE-1 and its tumorigenic variant 129Nu5002-1 Tu, became unresponsive to growth inhibition by transforming growth factor-beta. The only alteration uniquely correlated with the tumorigenic phenotype was loss of response to retinoic acid. This factor, which inhibits growth of normal and immortal but non-tumorigenic prostatic epithelial cells, had no effect on tumorigenic 129Nu5002-1 Tu cells. We previously reported that conversion of an SV40-immortalized prostatic epithelial cell line to tumorigenicity by introduction of the ras oncogene also resulted in loss of responsiveness to growth inhibitory activity of retinoic acid. 129Nu5002-1 Tu cells, which do not have an altered ras gene, gained the same phenotype. This suggests that loss of inhibition by retinoic acid may be a critical element in the tumorigenic conversion of prostatic epithelial cells.

    View details for Web of Science ID 000083973200008

    View details for PubMedID 10628324

  • Vitamin D and prostate cancer risk Symposium on Strategies for the Chemoprevention of Prostate Cancer Peehl, D. M. ELSEVIER SCIENCE BV. 1999: 392–94

    Abstract

    Prostate cancer is a progressive, multistep disease which presents many stages for intervention. Microscopic cancer is found in the prostate beginning by age 30 in about 20% of men, and the incidence increases steadily so that by the time a man is 90 years old, he has almost a 100% chance of having cancer in his prostate. Independent, multiple foci of cancer are present in the majority of prostate specimens, and the incidence of premalignant lesions is even higher than that of cancer. Yet, despite the high incidence of microscopic cancer, only 8% of men in the US present with clinically significant disease during their lifetime. Furthermore, only 3% of men in the US die of prostate cancer. In no other human cancer is there such disparity between the high incidence of microscopic malignancy and the relatively low death rate. Thus, there are many windows of opportunity for control of prostate cancer. Evidence from diverse areas of study - epidemiologic, molecular, genetic, cellular, animal models, and clinical trials - suggests that vitamin D may be an effective preventive agent against prostate cancer.

    View details for Web of Science ID 000080400800009

    View details for PubMedID 10325494

  • Induction of androgen receptor by 1 alpha,25-dihydroxyvitamin D-3 and 9-cis retinoic acid in LNCaP human prostate cancer cells ENDOCRINOLOGY Zhao, X. Y., Ly, L. H., Peehl, D. M., Feldman, D. 1999; 140 (3): 1205-1212

    Abstract

    We have recently shown that 1alpha,25-dihydroxyvitamin D3 [1,25-(OH)2D3] inhibits proliferation of LNCaP cells, an androgen-responsive human prostate cancer cell line. Also, 1,25-(OH)2D3 increases androgen receptor (AR) abundance and enhances cellular responses to androgen in these cells. In the current study, we have investigated the mechanism by which 1,25-(OH)2D3 regulates AR gene expression and the involvement of AR in the 1,25-(OH)2D3- and 9-cis retinoic acid (RA)-mediated growth inhibition of LNCaP cells. Northern blot analyses demonstrated that the steady-state messenger RNA (mRNA) level of AR was significantly increased by 1,25-(OH)2D3 in a dose-dependent manner. Time-course experiments revealed that the increase of AR mRNA by 1,25-(OH)2D3 exhibited delayed kinetics. In response to 1,25-(OH)2D3, AR mRNA levels were first detected to rise at 8 h and reached a maximal induction of 10-fold over the untreated control at 48 h; the effect was sustained at 72 h. Furthermore, the induction of AR mRNA by 1,25-(OH)2D3 was completely abolished by incubation of cells with cycloheximide, a protein synthesis inhibitor. 1,25-(OH)2D3 was unable to induce expression of an AR promoter-luciferase reporter. Together, these findings indicate that the stimulatory effect of 1,25-(OH)2D3 on AR gene expression is indirect. Western blot analyses showed an increase of AR protein in 1,25-(OH)2D3-treated cells. This increased expression of AR was followed by 1,25-(OH)2D3-induced inhibition of growth in LNCaP cells. Similar to 1,25-(OH)2D3, 9-cis RA also induced AR mRNA expression, and the effect of both hormones was additive. Moreover, 1,25-(OH)2D3 and 9-cis RA acted synergistically to inhibit LNCaP cell growth. These antiproliferative effects of 1,25-(OH)2D3 and 9-cis RA, alone or in combination, were blocked by the pure AR antagonist, Casodex. In conclusion, our results demonstrate that growth inhibition of LNCaP cells by 1,25-(OH)2D3 and 9-cis RA is mediated by an AR-dependent mechanism and preceded by the induction of AR gene expression. This finding, that differentiating agents such as vitamin D and A derivatives are potent inducers of AR, may have clinical implications in the treatment of prostate cancer.

    View details for Web of Science ID 000078733700025

    View details for PubMedID 10067845

  • Defined medium for normal adult human prostatic stromal cells IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY-ANIMAL Peehl, D. M., Sellers, R. G., Wong, S. T. 1998; 34 (7): 555-560

    Abstract

    Stromal-epithelial interactions are pivotal in many aspects of prostatic biology. A defined culture system is critical for the investigation of factors that regulate the growth and differentiation of human prostatic stromal cells. We have identified conditions which promote stromal cell attachment and proliferation in serum-free medium. MCDB 201, originally developed for the clonal growth of chick embryo fibroblasts, proved to be a superior basal medium of those that we tested. Supplementation of MCDB 201 with basic fibroblast growth factor (FGF), insulin-like growth factor (IGF), and platelet-derived growth factor (PDGF) permitted attachment and exponential growth of cells throughout a 7-d period with an initial inoculum as low as 10(3) cells per well of a 96-well microtiter dish. Using these assay conditions, we subsequently verified that basic FGF and IGF, but not PDGF, were required for optimal growth. No activity was found for heparin, transferrin, or the androgen R1881. Epidermal growth factor (EGF) didn't stimulate growth when added to medium containing basic FGF and IGF, but was moderately stimulatory when added to basal medium alone. Cholera toxin inhibited growth. This simple and efficient culture medium provides a suitable assay system for more extensive studies of growth regulation and differentiation of human prostatic stromal cells, and will provide the basis for future development of a defined medium that supports clonal growth. Characterization of stromal-epithelial interactions will be facilitated by the use of this defined culture system for stromal cells in conjunction with the serum-free culture systems previously developed for human prostatic epithelial cells.

    View details for Web of Science ID 000075277700011

    View details for PubMedID 9719415

  • Workgroup 2: Human xenograft models of prostate cancer PROSTATE STEARNS, M. E., Ware, J. L., Agus, D. B., Chang, C. J., FIDLER, I. J., Fife, R. S., Goode, R., Holmes, E., Kinch, M. S., Peehl, D. M., Pretlow, T. G., Thalmann, G. N. 1998; 36 (1): 56-58

    View details for Web of Science ID 000074349100009

    View details for PubMedID 9650917

  • Basic FGF, EGF, and PDGF modify TGF beta-induction of smooth muscle cell phenotype in human prostatic stromal cells PROSTATE Peehl, D. M., Sellers, R. G. 1998; 35 (2): 125-134

    Abstract

    We investigated the ability of a variety of growth factors to regulate the differentiation of prostatic fibroblasts into smooth muscle cells.Smooth muscle actin levels were monitored by immunoblot analysis and immunocytochemistry. Proliferation was measured in clonal growth assays and by cell counts.We determined that TGFbeta inhibited proliferation and induced smooth muscle differentiation of stromal cells derived from prostatic adenocarcinomas, as we previously reported for cells derived from the normal peripheral zone. Basic FGF, EGF, TGFalpha, and PDGF, but not IGF, retinoic acid, 1,25-dihydroxyvitamin D3, or androgen, attenuated induction of differentiation by TGFbeta, by a mechanism apparently unrelated to proliferation.Regulation of growth and differentiation occurs equivalently in prostatic stromal cells derived from adenocarcinomas and normal peripheral zone. TGFbeta is a potent inducer of the smooth muscle phenotype. Basic FGF, EGF and/or TGFalpha, and PDGF attenuate TGFbeta's activity, and promote a fibroblastic phenotype. Our studies provide an in vitro model system in which fibroblastic or smooth muscle cells can be promoted, maintained, and investigated in a defined manner. The results suggest that the ratio of fibroblasts to smooth muscle cells in the stroma reflects the relative levels of growth factors, which may be altered in diseased states.

    View details for Web of Science ID 000073180900006

    View details for PubMedID 9568676

  • Expression of prostatic factors measured by reverse transcription polymerase chain reaction in human papillomavirus type 18 deoxyribonucleic acid immortalized prostate cell lines UROLOGY Weijerman, P. C., Zhang, Y., Shen, J. C., Dubbink, H. J., Romijn, J. C., Peehl, D. M., Schroder, F. H. 1998; 51 (4): 657-662

    Abstract

    To investigate expression of the prostatic markers prostate-specific antigen (PSA), prostate-specific membrane antigen (PSM), and the androgen receptor (AR) after human papillomavirus (HPV) type 18 deoxyribonucleic acid (DNA) transfection and subsequent immortalization of human prostate epithelial cells.Recently, two human prostate epithelial cell lines were established by HPV transformation: PZ-HPV-7, derived from normal peripheral zone (PZ) tissue, and CA-HPV-10, derived from high Gleason grade adenocarcinoma. Expression of PSA was studied by the reverse transcription polymerase chain reaction (RT-PCR), because in preliminary studies using immunocytochemistry and Northern blotting, no PSA expression was found. PSM was analyzed by RT-PCR and nested RT-PCR. These analyses included primary human prostate cell strains. Furthermore, androgen-supplemented methylthiazol tetrazolium (MTT) growth assays were performed and expression of AR was studied by immunocytochemistry. Prostate carcinoma cell lines LNCaP and PC-346C were included as positive controls and breast carcinoma cell line MCF-7 as a negative control.Both cell lines exhibited low levels of RNA for PSA and PSM in comparison with cell lines LNCaP and PC-346C. AR expression by immunocytochemistry was negative using monoclonal antibody F39.4 and polyclonal antibody SP-197. In an androgen-supplemented environment, growth rates of both HPV immortalized cell lines were not stimulated in contrast to LNCaP.RNA transcripts of PSA and PSM were detected by RT-PCR in HPV immortalized prostate epithelial cell lines PZ-HPV-7 and CA-HPV-10. The expression of prostate-specific markers may further validate the utility of this stepwise transformation model of human prostate carcinogenesis.

    View details for Web of Science ID 000073274300028

    View details for PubMedID 9586625

  • Expression and characterization of recombinant type 2 3 alpha hydroxysteroid dehydrogenase (HSD) from human prostate: Demonstration of bifunctional 3 alpha/17 beta-HSD activity and cellular distribution MOLECULAR ENDOCRINOLOGY Lin, H. K., Jez, J. M., Schlegel, B. P., Peehl, D. M., Pachter, J. A., Penning, T. M. 1997; 11 (13): 1971-1984

    Abstract

    In androgen target tissues, 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD) may regulate occupancy of the androgen receptor (AR) by catalyzing the interconversion of 5alpha-dihydrotestosterone (5alpha-DHT) (a potent androgen) and 3alpha-androstanediol (a weak androgen). In this study, a 3alpha-HSD cDNA (1170 bp) was isolated from a human prostate cDNA library. The human prostatic 3alpha-HSD cDNA encodes a 323-amino acid protein with 69.9%, 84.1%, 99.4%, and 87.9% sequence identity to rat liver 3alpha-HSD and human type 1, type 2, and type 3 3alpha-HSDs, respectively, and is a member of the aldo-keto reductase superfamily. The close homology with human type 2 3alpha-HSD suggests that it is either identical to this enzyme or a structural allele. Surprisingly, when the recombinant protein was expressed and purified from Escherichia coli, the enzyme did not oxidize androsterone when measured spectrophotometrically, an activity previously assigned to recombinant type 2 3alpha-HSD using this assay. Complete kinetic characterization of the purified protein using spectrophotometric, fluorometric, and radiometric assays showed that the catalytic efficiency favored 3alpha-androstanediol oxidation over 5alpha-DHT reduction. Using [14C]-5alpha-DHT as substrate, TLC analysis confirmed that the reaction product was [14C]-3alpha-androstanediol. However, in the reverse reaction, [3H]-3alpha-androstanediol was oxidized first to [3H]-androsterone and then to [3H]-androstanedione, revealing that the expressed protein possessed both 3alpha- and 17beta-HSD activities. The 17beta-HSD activity accounted for the higher catalytic efficiency observed with 3alpha-androstanediol. These findings indicate that, in the prostate, type 2 3alpha-HSD does not interconvert 5alpha-DHT and 3alpha-androstanediol but inactivates 5alpha-DHT through its 3-ketosteroid reductase activity. Levels of 3alpha-HSD mRNA were measured in primary cultures of human prostatic cells and were higher in epithelial cells than stromal cells. In addition, elevated levels of 3alpha-HSD mRNA were observed in epithelial cells derived from benign prostatic hyperplasia and prostate carcinoma tissues. Expression of 3alpha-HSD was not prostate specific, since high levels of mRNA were also found in liver, small intestine, colon, lung, and kidney. This study is the first complete characterization of recombinant type 2 3alpha-HSD demonstrating dual activity and cellular distribution in the human prostate.

    View details for Web of Science ID A1997YK06500009

    View details for PubMedID 9415401

  • Selection of human cervical epithelial cells that possess reduced apoptotic potential to low-oxygen conditions CANCER RESEARCH Kim, C. Y., Tsai, M. H., OSMANIAN, C., Graeber, T. G., Lee, J. E., Giffard, R. G., DiPaolo, J. A., Peehl, D. M., Giaccia, A. J. 1997; 57 (19): 4200-4204

    Abstract

    Since human papillomavirus (HPV) infection is strongly associated with cervical neoplasia and tumor hypoxia has prognostic significance in human cervical carcinomas, we examined the relationship between hypoxia and apoptosis in human cervical epithelial cells expressing high-risk HPV type 16 oncoproteins. In vitro, hypoxia stimulated both p53 induction and apoptosis in primary cervical epithelial cells infected with the HPV E6 and E7 genes but not in cervical fibroblasts infected with E6 and E7. Furthermore, cell lines derived from HPV-associated human cervical squamous cell carcinomas were substantially less sensitive to apoptosis induced by hypoxia, indicating that these cell lines have acquired additional genetic alterations that reduced their apoptotic sensitivity. Although the process of long-term cell culturing resulted in selection for subpopulations of HPV oncoprotein-expressing cervical epithelial cells with diminished apoptotic potential, the exposure of cells to hypoxia greatly accelerated the selection process. These results provide evidence for the role of hypoxia-mediated selection of cells with diminished apoptotic potential in the progression of human tumors and can in part explain why cervical tumors that possess low pO2 values are more aggressive.

    View details for PubMedID 9331075

  • 1 alpha 25-dihydroxyvitamin D-3 actions in LNCaP human prostate cancer cells are androgen-dependent ENDOCRINOLOGY Zhao, X. Y., Ly, L. H., Peehl, D. M., Feldman, D. 1997; 138 (8): 3290-3298

    Abstract

    We and others have recently shown that 1alpha,25-dihydroxyvitamin D3 [1,25-(OH)2D3] significantly inhibits cell proliferation and increases secretion of prostate-specific antigen (PSA) in LNCaP cells, an androgen-responsive human prostate cancer cell line. The present study was designed to investigate the possible interactions between 1,25-(OH)2D3 and androgens in the regulation of LNCaP cellular function. LNCaP cell growth was dose-dependently inhibited by 1,25-(OH)2D3 (60% inhibition at 10 nM) when cells were cultured in medium supplemented with FBS (FBS medium). 1,25-(OH)2D3-treated cells showed a 5-fold increase in PSA secretion, similar to the increase seen in dihydrotestosterone (DHT)-treated cells. In combination, 1,25-(OH)2D3 and DHT synergistically enhanced PSA secretion 22-fold. This synergistic effect was even greater when cells were cultured in medium supplemented with charcoal-stripped serum (CSS medium), where endogenous steroids are substantially depleted. Under these conditions, 1,25-(OH)2D3 and DHT together stimulated PSA secretion up to 50-fold over the untreated control. Radioligand binding assays and Western blot analyses showed that the androgen receptor (AR) content was increased significantly by 1,25-(OH)2D3 at 48 h. Furthermore, the steady-state mRNA level of AR was up-regulated approximately 2-fold by 1,25-(OH)2D3 at 24 h. When cells were grown in CSS medium, 1,25-(OH)2D3 alone no longer inhibited cell growth or induced PSA secretion. Titration experiments revealed that the addition of DHT at 1 nM to the medium restored the antiproliferative activity of 1,25-(OH)2D3. Conversely, an antiandrogen, Casodex, completely blocked 1,25-(OH)2D3 antiproliferative and PSA stimulation activities when cells were cultured in FBS medium. In conclusion, these results demonstrate that the antiproliferative and PSA induction activities of 1,25-(OH)2D3 in LNCaP cells are dependent upon androgen action and that AR up-regulation by 1,25-(OH)2D3 likely contributes to the synergistic actions of 1,25-(OH)2D3 and DHT in these cells.

    View details for Web of Science ID A1997XL84200028

    View details for PubMedID 9231780

  • 1 alpha,25-dihydroxyvitamin D-3 actions in LNCaP human prostate cancer cells are androgen-dependent. Zhao, X. Y., Ly, L. H., Peehl, D. M., Feldman, D. AMER SOC BONE & MINERAL RES. 1997: F689–F689
  • Loss of response to epidermal growth factor and retinoic acid accompanies the transformation of human prostatic epithelial cells to tumorigenicity with v-Ki-ras CARCINOGENESIS Peehl, D. M., Wong, S. T., Sellers, R. G., Jin, S., Rhim, J. S. 1997; 18 (8): 1643-1650

    Abstract

    Growth factor-independent proliferation and loss of response to differentiation factors are believed to be critical elements in carcinogenesis. We have developed an in vitro model of human prostatic carcinogenesis by the introduction of SV40 DNA into normal prostatic epithelial cells to create a transformed, immortal cell line, pRNS-1-1. This non-tumorigenic cell line responded similarly to normal prostatic epithelial cells to most growth- and differentiation-regulatory factors, with the notable exception of loss of response to the inhibitory factor 1,25-dihydroxyvitamin D3. In this study, we describe the introduction of the ras oncogene into pRNS-1-1 cells to create a tumorigenic cell line, pRNS-1-1/ras. In addition to an attenuated response to 1,25-dihydroxyvitamin D3, these cells also became unresponsive to retinoic acid and gained the ability to undergo clonal proliferation in the absence of epidermal growth factor (EGF). EGF-independent growth could not be linked to the production of autocrine transforming growth factor-alpha, but instead was likely due to sustained signaling by the ras oncogene, bypassing ligand-activation of the EGF receptor. Ligand-independent proliferation, coupled with the loss of response to the growth-inhibitory and differentiation agent retinoic acid, may be important elements in the conversion of human prostatic epithelial cells to tumorigenicity.

    View details for Web of Science ID A1997XQ87800030

    View details for PubMedID 9276642

  • Human papillomavirus detection by polymerase chain reaction in benign and malignant prostate tissue is dependent on the primer set utilized 91st Annual Meeting of the American-Urological-Association Terris, M. K., Peehl, D. M. CAHNERS PUBL CO. 1997: 150–56

    Abstract

    Prior investigations evaluating the presence of human papillomavirus (HPV) in prostatic tissue by polymerase chain reaction (PCR) technology have yielded detection rates of 0% to 100%. Contamination by viral DNA from prostatic urethral colonization or less than optimal laboratory conditions have been suggested to explain this discrepancy. In addition, the various investigations have differed in the specific oligonucleotide primers utilized for amplification and, therefore, have searched for different segments of the viral genome. The objective of this study is to address these differences.Forty-one archival radical prostatectomy specimens were evaluated, identifying areas of normal and abnormal histology. Meticulous technique was used during tissue acquisition, histologic confirmation, DNA isolation, PCR amplification, polyacrylamide gel electrophoresis, and staining. Primers for a 126- and 99-base pair (bp) fragment of the E6 portion of HPV 16 as well as a consensus primer for the L1 portion of the papillomavirus genome were utilized.Of the normal prostatic tissues, 13.5% (5/37) contained the 126-bp E6 viral DNA as did 33.3% (7/21) of benign prostatic hyperplasia (BPH) samples, 25% (5/20) of dysplasia, 18.2% (2/11) of Gleason grades 1 and 2 adenocarcinoma, 25.9% (7/27) of Gleason grade 3 adenocarcinoma, and 6.7% (1/15) of Gleason grade 4 adenocarcinoma. Sections from the urethras of the prostatectomy specimens contained viral DNA in 31.7% (13/41). Viral detection was variable among different specimens in the same patient. With amplification for the 99-bp fragment of HPV 16, 1 of 37 normal (2.7%), 2 of 21 BPH (9.5%), 1 of 20 dysplasia (5.0%), and 2 of 53 cancer (3.8%) specimens revealed HPV DNA. In none of the specimens was DNA amplified using primers for a 450-bp fragment of the L1 portion of HPV.Previously published discrepancies in HPV detection may be solely due to the differences in primer sets utilized.

    View details for Web of Science ID A1997XJ07600032

    View details for PubMedID 9218040

  • Decreased expression of Wilms' tumor gene WT-1 and elevated expression of insulin growth factor-II (IGF-II) and type 1 IGF receptor genes in prostatic stromal cells from patients with benign prostatic hyperplasia JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Dong, G. Y., Rajah, R., Vu, T., Hoffman, A. R., Rosenfeld, R. G., Roberts, C. T., Peehl, D. M., Cohen, P. 1997; 82 (7): 2198-2203

    Abstract

    Benign prostatic hyperplasia (BPH) is a common proliferative disorder of unknown etiology. We have previously documented that the insulin-like growth factor (IGF) axis is critical for prostate cell growth and is abnormal in BPH. The type 1 IGF receptor (IGF-1R) is constitutively expressed by most body tissues and plays a significant role in regulating cell proliferation, consistent with the role of its ligands (IGF-I and IGF-II) as important mitogenic factors. The Wilms' tumor gene product (WT-1) is a tumor suppressor that has been shown to be altered in rare kidney tumors and is known to regulate IGF-II and IGF-1R. We investigated the possibility that the expression of prostatic WT-1, IGF-1R, and IGF-II genes is altered in patients with BPH. We utilized primary cultures of prostatic stromal cells grown from normal (n = 9) and hyperplastic (n = 9) surgical specimens and analyzed WT-1, IGF-1R, and IGF-II messenger RNA levels. In all of the BPH cell strains, WT-1 expression (measured by RT-PCR and RNase protection assays) was strikingly lower than that found in normal strains (0-20% of normal, mean 14% of normal, P < 0.01). The expression of both the IGF-1R (300% of normal, P < 0.05) and IGF-II (1000% of normal, P < 0.01) messenger RNAs was higher in BPH strains as compared with normal strains. No changes were seen in stromal cell strains derived from prostatic adenocarcinoma. Thus, in cultured human prostatic stromal cell strains from patients with BPH, decreased WT-1 gene expression is associated with increases in the expression of the IGF-1R and IGF-II genes that are known transcriptional targets of WT-1. These findings indicate that reduced expression of the WT-1 tumor suppressor gene and elevated IGF-1R and IGF-II gene expression may be involved in the pathophysiology of prostatic hyperplasia, implying a new role for the Wilms' tumor gene in nonmalignant states.

    View details for Web of Science ID A1997XH82400030

    View details for PubMedID 9215294

  • Induction of smooth muscle cell phenotype in cultured human prostatic stromal cells EXPERIMENTAL CELL RESEARCH Peehl, D. M., Sellers, R. G. 1997; 232 (2): 208-215

    Abstract

    Stromal cells are key regulators of growth and differentiation in the adult human prostate. Alterations in the stroma are believed to initiate the development of benign prostatic hyperplasia, and stromal-epithelial interactions may have a role in malignant progression. The prostatic stroma is composed of two major cell types, smooth muscle cells and fibroblasts. Cell cultures from the prostatic stroma have been established by several investigators, but the phenotype of these cells has not been extensively characterized and it is not clear whether they are fibroblastic or smooth muscle-like. In this study, the response of stromal cells cultured from normal prostatic tissues to transforming growth factor-beta (TGF beta) was investigated. We confirmed a previous report that TGF beta inhibited the growth of prostatic stromal cells in serum-containing medium, and showed that inhibition also occurred in serum-free medium. Growth inhibition by TGF beta was irreversible after 24 to 72 h of exposure. In the absence of TGF beta, cells were fibroblastic and expressed vimentin and fibronectin but little alpha-smooth muscle actin. After 3 days of exposure to 1 ng/ml of TGF beta, the majority of cells expressed alpha-smooth muscle actin and desmin, as demonstrated by immunocytochemistry and immunoblot analysis. This effect was specific and alpha-smooth muscle actin was not induced by two other growth-inhibitory factors, retinoic acid or 1,25-dihydroxyvitamin D3. These results suggest that TGF beta is an important regulator of growth and differentiation of prostatic stromal cells and that a smooth muscle cell phenotype is promoted in the presence of TGF beta.

    View details for Web of Science ID A1997WX48900002

    View details for PubMedID 9168795

  • Acid-activated insulin-like growth factor binding protein protease activity of cathepsin D in normal and malignant prostatic epithelial cells and seminal plasma JOURNAL OF CELLULAR PHYSIOLOGY Nunn, S. E., Peehl, D. M., Cohen, P. 1997; 171 (2): 196-204

    Abstract

    In this study, we demonstrate insulin-like growth factor binding protein (IGFBP) acid proteolysis in conditioned media (CM) from normal and malignant primary cultures of prostatic epithelial cells, prostatic cell lines, and in seminal plasma. We further demonstrate the absence of such activity in CM from prostatic stromal cells. Radio-labeled IGFBPs (1-6) were incubated with various acidified CM and seminal plasma. None of these media showed IGFBP proteolytic activity at neutral pH, but all CM from prostatic epithelial cells (PC-E) demonstrated strong IGFBP proteolysis at acidic pH. No acid-activated proteolysis was observed in the CM from stromal cell cultures. In order to ascertain the role of cathepsin D, anti-cathepsin antibodies were used to immunodeplete the media of the selected enzymes prior to incubation with IGFBPs. Depletion of cathepsin D greatly reduced the proteolytic activity of the PC-E CM. Additionally, purified cathepsin D yielded a digestion pattern identical to that produced by prostatic cell CM and seminal plasma, following acidic incubation with IGFBP-3. Remarkably, the proteolytic pattern generated by seminal plasma, when incubated with IGFBP-3 at neutral pH, corresponded to that produced by prostate-specific antigen (PSA), demonstrating the interpolation of both neutral and acid proteases from prostate cells into seminal plasma. In conclusion, prostatic epithelial cells secrete acid-specific IGFBP protease(s) related to cathepsin D. Although no significant statistical difference was observed in the degree of acid-specific proteolysis in the media from normal versus malignant primary epithelial cell cultures, physiological characteristics of the malignant state might facilitate increased cathepsin D activity. We suspect this proteolysis may play a role in prostatic cell proliferation and invasive tumor growth.

    View details for Web of Science ID A1997WW03300010

    View details for PubMedID 9130467

  • Parathyroid hormone-related protein is not an autocrine growth factor for normal prostatic epithelial cells PROSTATE Peehl, D. M., Edgar, M. G., Cramer, S. D., Deftos, L. J. 1997; 31 (1): 47-52

    Abstract

    Parathyroid hormone-related protein (PTHrP) is the primary factor responsible for humoral hypercalcemia of malignancy. The hypercalcemic actions of PTHrP occur via stimulation of renal distal tubular calcium reabsorption and increased osteoclastic bone resorption. These effects of PTHrP are thought to be mediated through a common parathyroid hormone (PTH)/PTHrP receptor. In addition to the well-established role of PTHrP in bone remodeling, PTHrP is believed to be an important mediator of cellular growth and differentiation in a number of nonbony tissues. We recently demonstrated abundant expression of PTHrP in normal and malignant human prostatic tissues, and in cultured prostatic epithelial cells.In vitro assays were used to test growth-regulatory activity of synthetic and endogenous PTHrP peptides on normal prostatic epithelial cells.No growth-regulatory activity could be demonstrated.PTHrP is not an autocrine growth factor for normal prostatic epithelial cells.

    View details for Web of Science ID A1997WT87500008

    View details for PubMedID 9108886

  • The role of hypoxia, p53, and apoptosis in human cervical carcinoma pathogenesis Kim, C. Y., Tsai, M. H., OSMANIAN, C., CALKINS, D. P., Graeber, T. G., Greenspan, D. L., Kennedy, A. S., Rinker, L. H., Varia, M. A., DiPaolo, J. A., Peehl, D. M., Raleigh, J. A., Giaccia, A. J. ELSEVIER SCIENCE INC. 1997: 206–
  • Prostate specific antigen cleaves parathyroid hormone-related protein in the PTH-like domain: Inactivation of PTHrP-stimulated cAMP accumulation in mouse osteoblasts JOURNAL OF UROLOGY Cramer, S. D., Chen, Z. X., Peehl, D. M. 1996; 156 (2): 526-531

    Abstract

    To determine whether parathyroid hormone-related protein (PTHrP) is a substrate of prostate-specific antigen (PSA) and how the biological activity of PTHrP may be altered by cleavage with PSA.Prostate-specific antigen cleavage of recombinant human PTHrP 1-141 was conducted in vitro at 37C and analyzed by SDS-PAGE. Five rounds of automated amino-terminal amino acid sequence analysis were performed on blotted PSA-cleaved PTHrP peptide fragments to determine the PSA cleavage sites. The mouse osteoblast cell line MC3T3-E1 was used to test whether PSA cleavage of PTHrP 1-141 altered its ability to stimulate cAMP production.Prostate-specific antigen was found to specifically cleave PTHrP 1-141 in a time- and dose-dependent manner. Cleavage of PTHrP 1-141 by PSA generated fragments on Coomassie-stained acrylamide gels that migrated with mobilities that corresponded to 19.5, 17, 15 and < 7 kd. The preferred PSA cleavage site of PTHrP 1-141 was determined to be at the carboxyl-terminus of phenylalanine 23, consistent with chymotryptic-like enzymatic activity of PSA. Cleavage of PTHrP by PSA completely abolished the ability of PTHrP to stimulate cAMP production.Cleavage of PTHrP 1-141 by PSA carboxyl-terminal to phenylalanine 23 represents a unique pattern of PTHrP processing that may be specific to the prostate. Prostate-specific antigen inactivation of the cAMP-inducing activity of PTHrP 1-141 demonstrates that PSA cleavage regulates the biological activity of PTHrP. These results have implications for the role of PTHrP in prostate cancer metastasis to bone and its subsequent regulation of bone remodeling. Study of the biological activities of the PSA-generated PTHrP peptides identified in this study merits further investigation.

    View details for Web of Science ID A1996UX15200074

    View details for PubMedID 8683730

  • Parathyroid hormone-related protein (PTHrP) is an epidermal growth factor-regulated secretory product of human prostatic epithelial cells PROSTATE Cramer, S. D., Peehl, D. M., Edgar, M. G., Wong, S. T., Deftos, L. J., Feldman, D. 1996; 29 (1): 20-29

    Abstract

    Parathyroid hormone-related protein (PTHrP) has previously been shown to be expressed in human prostatic tissue and in prostatic cancer cell lines. In the present study, PTHrP immunoreactivity was detected in the glandular epithelium of normal prostate and benign prostatic hyperplasia (BPH), as well as in prostatic adenocarcinoma (CaP). Epithelial cell cultures derived from normal, BPH, and CaP tissues were also stained by antibodies against PTHrP, and northern analysis revealed multiple transcripts of PTHrP in the cellular RNA. PTHrP (1-34) was measurable by radioimmunoassay (RIA) in media conditioned by the prostatic epithelial cell cultures, and PTHrP accumulated in conditioned media during a 72 hr time course. Addition of complete growth medium to starved cells resulted in increased PTHrP mRNA levels by 1 hr, with maximal stimulation at 8-24 hr. Several individual factors contained in the complete growth medium were tested for their ability to regulate PTHrP expression. Epidermal growth factor (EGF) was the major inducer of PTHrP expression, while cholera toxin, bovine pituitary extract, hydrocortisone, and insulin had minimal or no effect on PTHrP transcript levels. Since each of these factors is growth stimulatory, the unique ability of EGF to induce PTHrP is apparently unrelated to mitogenicity. 1,25-Dihydroxyvitamin D3[1,25(OH)2D3], an inhibitor of PTHrP expression in several other cell types, had no effect on steady-state levels of PTHrP mRNA expressed by epithelial cells in complete growth medium, although prostate cells have vitamin D receptors and are responsive to 1,25(OH)2D3 in other ways. Our results indicate that PTHrP expression is not confined to the neuroendocrine cells of the human prostate and that our culture system can be used as a model to investigate the role of PTHrP in the prostate.

    View details for Web of Science ID A1996UX88000003

    View details for PubMedID 8685051

  • 7S nerve growth factor is an insulin-like growth factor-binding protein protease ENDOCRINOLOGY Rajah, R., Bhala, A., Nunn, S. E., Peehl, D. M., Cohen, P. 1996; 137 (7): 2676-2682

    Abstract

    Insulin-like growth factor (IGF)-binding protein (IGFBP) proteases modulate IGF action by cleaving IGFBPs into fragments with lower affinity to IGFs, thereby increasing the levels of free IGFs. We have previously documented that prostate-specific antigen (PSA), a serine protease of the kallikrein family, is an IGFBP-3 protease. In this study, we characterized the potential IGFBP proteolytic activity of nerve growth factor (NGF gamma-subunit), which shares high sequence homology with PSA. [125I]IGFBP-3 was cleaved by NGF (but not other kallikreins) at a 3-fold lower concentration than that of PSA, thus proving NGF to be a more potent IGFBP protease than PSA. NGF-generated, lower mol wt IGFBP-3 fragments, detected by immunoblotting and cross-linking to IGFs, had a lower affinity to IGFs than intact IGFBP-3. Unlike PSA, which cleaves primarily IGFBP-3 and -5, NGF also displayed potent proteolytic activity against IGFBP-4 and -6. These data suggest that NGF may be involved in the growth of cells by more than one mechanism. In addition to binding to its receptors, NGF is capable of cleaving IGFBPs and, thus, enhancing IGF action. This synergistic action between NGF and IGF may have important implications on cell growth, development, and repair in the brain and other tissues.

    View details for Web of Science ID A1996UR95500006

    View details for PubMedID 8770886

  • Keratin 19 in the adult human prostate: Tissue and cell culture studies CELL AND TISSUE RESEARCH Peehl, D. M., Sellers, R. G., McNeal, J. E. 1996; 285 (1): 171-176

    Abstract

    Keratin 19 is found primarily in simple epithelia. In mammary epithelia, keratin 19 was localized to a subset of luminal cells, suggesting that keratin 19-negative cells may be the proliferative compartment of the secretory cell lineage. The structural and functional similarities of prostate and breast led us to examine keratin 19 expression in the prostate. Immunohistochemical studies revealed that keratin 19 expression was heterogeneous and frequently occurred in basal as well as in luminal cells of normal, dysplastic, and benign hyperplastic tissues. Keratin 19 was observed in cancer, but usually in a minority of cells. This was in dramatic contrast to invasive breast cancers, which are reportedly uniformly positive for keratin 19. Prostatic epithelial cells cultured from tissues of all histologies expressed keratin 19. In summary, keratin 19 does not obviously correlate with any epithelial cell lineage or phenotype in the prostate.

    View details for Web of Science ID A1996UW98300020

    View details for PubMedID 8766871

  • Recent advances in the development of animal and cell culture models for prostate cancer research A minireview. Urologic oncology Bosland, M. C., Chung, L. W., Greenberg, N. M., Ho, S. M., Isaacs, J. T., Lane, K., Peehl, D. M., Thompson, T. C., van Steenbrugge, G. J., van Weerden, W. M. 1996; 2 (4): 99

    View details for PubMedID 21224146

  • The role of the Sertoli cell in active immunosuppression in the human testis BRITISH JOURNAL OF UROLOGY Turek, P. J., Malkowicz, S. B., Tomaszewski, J. E., WEIN, A. J., Peehl, D. 1996; 77 (6): 891-895

    Abstract

    To define the involvement of Sertoli cells in immune suppression within the testis by identifying surface receptor molecules that describe or communicate with cells of the immune system.Ten pairs of human testes obtained from orchidectomy were stained immunohistochemically with established monoclonal antibodies that identify common cells of the immune system.Patterns of staining were similar in all testes. Class I major histocompatibility complex (MHC) antigen, important for self/non-self recognition, was found on Sertoli cells. Class II MHC antigen, important for immune cell interaction and limited to immune cells, was absent. Leu M3, characteristically found on macrophages, was also seen on Sertoli cells but no receptor antigens defining basic T cell types (CD4, CD8), B cells (Leu 12) or natural killer cells (Leu 11) were seen on Sertoli cells.The Sertoli cell membrane lacks lymphocyte-cell surface markers but harbours somatic cell and macrophage markers. This suggests that immunosuppression within the testis is not maintained through classic lymphocyte receptors. The presence of a macrophage marker is consistent with the known phagocytic activity of the Sertoli cell.

    View details for Web of Science ID A1996UP44100022

    View details for PubMedID 8705228

  • KGF and EGF differentially regulate the phenotype of prostatic epithelial cells GROWTH REGULATION Peehl, D. M., Wong, S. T., Rubin, J. S. 1996; 6 (1): 22-31

    Abstract

    Previous studies indicate that keratinocyte growth factor (KGF) acts as a paracrine factor in the prostatic epithelium and epidermal growth factor (EGF) acts as an autocrine factor. In serum-free medium, KGF or EGF promoted similar growth of human prostatic epithelial cells. Response to two growth-inhibitory factors (suramin and transforming growth factor-beta), and expression of keratins and prostate-specific antigen (PSA), were similar with either mitogen. However, colonies in medium with KGF were very compact with extensive intercellular bonds, whereas colonies with EGF consisted of widely-separated cells. Growth was decreased to a greater extent by deletion of growth factors from medium with KGF versus EGF, and retinoic acid was 10-fold more potent at inducing growth inhibition and differentiation-associated keratin with KGF compared with EGF. We conclude that regulation of growth and differentiation in the prostate might vary depending on the availability of KGF versus EGF.

    View details for Web of Science ID A1996UK51000004

    View details for PubMedID 8717447

  • The role of insulin-like growth factors in prostate biology JOURNAL OF ANDROLOGY Peehl, D. M., Cohen, P., Rosenfeld, R. G. 1996; 17 (1): 2-4

    View details for Web of Science ID A1996UA10800002

    View details for PubMedID 8833734

  • Cellular biology of prostatic growth factors International Conference on Androgenic Hormones, Prostate Cancer, and Benign Prostatic Hyperplasia Peehl, D. M. WILEY-BLACKWELL. 1996: 74–78

    Abstract

    The etiology of prostate cancer or of benign prostatic hyperplasia (BPH) is essentially not understood. It is becoming clear, however, that major determinants of the malignant or hyperplastic phenotype are various growth-stimulatory or -inhibitory factors and their receptors, whose inappropriate expression or loss disrupts normal regulation of cell proliferation and differentiation. Cell culture has been a versatile tool for studying the expression and interaction of growth factors in prostatic cells. Immunohistochemistry and in situ hybridization have provided a view of growth factor expression coupled with histopathology. The eventual definition of autocrine, paracrine, and endocrine pathways of growth regulation in the human prostate will facilitate the design of new preventative, diagnostic, and therapeutic strategies.

    View details for Web of Science ID A1996UB91100014

    View details for PubMedID 8630234

  • Simian virus 40-, but not human papillomavirus-, transformation of prostatic epithelial cells results in loss of growth-inhibition by 1,25-dihydroxyvitamin D-3 INTERNATIONAL JOURNAL OF ONCOLOGY Gross, C., Skowronski, R. J., Plymate, S. R., Rhim, J. S., Peehl, D. M., Feldman, D. 1996; 8 (1): 41-47

    Abstract

    In addition to its well known calcemic actions, 1,25-dihydroxyvitamin D-3 [1,25(OH)(2)D] exhibits differentiating and antiproliferative effects in several types of cancer cells. 1,25(OH)(2)D receptors (VDR) as well as 1,25(OH)(2)D-mediated growth-inhibition have been demonstrated in human prostate cancer cell lines. In order to further develop model systems for the study of 1,25(OH)(2)D action and to elucidate the mechanism of growth-inhibition, we studied several human prostate cell lines immortalized with either simian virus 40 (SV40) or human papillomavirus type 18 (HPV). The SV40-transformed cell lines P69SV40-T and P153SV40-T were not growth-inhibited by 1,25(OH)(2)D at concentrations as high as 100 nM, whereas the HPV-transformed cells PZ-HPV-7 and CA-HPV-10 were growth-inhibited. All cell lines expressed VDR, and VDR mRNA was demonstrated by Northern blot analysis. All cells exhibited induction of 24-hydroxylase mRNA, a 1,25(OH)(2)D responsive gene, after 1,25(OH)(2)D treatment. In an attempt to understand the apparent dissociation of 1,25(OH)(2)D actions in the SV40-transformed cells, we turned to the human prostate cancer cell line DU 145. These cells, like the SV40-transformed cells, are not growth-inhibited but demonstrate induction of 24-hydroxylase mRNA after 1,25(OH)(2)D treatment. DU 145 cells contain a mutated retinoblastoma gene (Rb) which contributes to their uncontrolled growth, analogous to the disruption of Rb by SV40 and HPV. We compared DU,145 cells to DU 145 cells transfected with normal Rb (DU 145/Rb). Similar to DU 145, DU 145/Rb cells were not growth-inhibited by 1,25(OH)(2)D, while 24-hydroxylase mRNA was induced. These results suggest that divergent pathways mediate the growth-inhibitory effect of 1,25(OH)(2)D and its induction of 24-hydroxylase. It also appears that the antiproliferative effect of 1,25(OH)(2)D is mediated by an Rb-independent mechanism.

    View details for Web of Science ID A1996TM02400006

    View details for PubMedID 21544329

  • KERATINOCYTE GROWTH-FACTOR - AN ANDROGEN-REGULATED MEDIATOR OF STROMAL EPITHELIAL INTERACTIONS IN THE PROSTATE WORLD JOURNAL OF UROLOGY Peehl, D. M., Rubin, J. S. 1995; 13 (5): 312-317

    Abstract

    Members of the fibroblast growth factor (FGF) family are important growth-regulatory elements. Of the FGFs, keratinocyte growth factor (KGF) appears to have unique properties that implicate it as a paracrine factor in the prostate. Two KGF transcripts (approximately to 2.4 and 5.0 kb) encode a protein of approximately 22 kDa. In contrast to several other members of the FGF family, KGF has a signal peptide and is actively secreted. Cellular response to KGF is mediated by a specific receptor that is transcribed from an alternately spliced variant of the FGF type 2 receptor (FGFR-2). KGF transcripts have been detected in prostatic tissues and in stromal cells cultured from rat and human prostates as well as in a variety of stromal cells derived from other organs. Prostatic epithelial cells and numerous other types of epithelial cells are targets of KGF's mitogenic activity. Several factors involved in wound healing regulate the expression of KGF, but androgen regulation of KGF is of greatest relevance to the role of KGF in the prostate. Current efforts to localize and manipulate KGF activity in vivo should reveal the significance of KGF expression and function in the prostate and in other organs.

    View details for Web of Science ID A1995TC93300008

    View details for PubMedID 8581003

  • THE INSULIN-LIKE GROWTH-FACTOR SYSTEM IN THE PROSTATE WORLD JOURNAL OF UROLOGY Peehl, D. M., Cohen, P., Rosenfeld, R. G. 1995; 13 (5): 306-311

    Abstract

    The insulin-like growth factor (IGF) system is involved in the regulation of cell growth. The system involves a network of molecules that includes the IGFs themselves (IGF-I and -II), IGF receptors (types I and II), IGF-binding proteins (IGFBP-1 through -6), and IGFBP proteases. Characterization of this complex system in the prostate has recently been initiated. Prostatic cell lines as well as primary cultures of prostatic epithelial and stromal cells have been analyzed for expression of IGFs, receptors, and IGFBPs. Prostatic epithelial cells and, quite likely, stromal cells as well respond to the mitogenic activity of IGFs via the type I IGF receptor. Prostatic stromal cells synthesize and secrete IGF-II; there is evidence that prostatic cell lines also synthesize IGFs, but this has not been confirmed in primary cultures of prostatic epithelial cells. Prostatic stromal and epithelial cells secrete a number of IGFBPs. The biological impact of some of these IGFBPs on the growth of prostatic cells has been examined, and proteolytic cleavage of IGFBP-3 by prostate-specific antigen (PSA) has been demonstrated. Aberrations in several elements of the IGF system have been observed in stromal cells derived from benign prostatic hyperplasia (BPH). The IGF system may therefore have a part in the etiology of BPH as well as in normal and malignant processes in the prostate.

    View details for Web of Science ID A1995TC93300007

    View details for PubMedID 8581002

  • Suramin, hydrocortisone, and retinoic acid modify inhibitory effects of 1,25-dihydroxyvitamin D(3) on prostatic epithelial cells. Urologic oncology Peehl, D. M., Wong, S. T., Cramer, S. D., Gross, C., Feldman, D. 1995; 1 (5): 188-194

    Abstract

    The proliferation of prostatic epithelial cells is regulated by the complex interplay of numerous growth-stimulatory and growth-inhibitory factors. 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] has recently been identified as a potent inhibitor of the growth of prostatic epithelial cells. Epidemiologic studies indicate that vitamin D deficiency may be a risk factor for the development of clinical prostate cancer, possibly due to increased growth and reduced differentiation of prostatic cells in an environment with decreased 1,25(OH)(2)D(3). The application of vitamin D or analogs in chemotherapy against prostate and other cancers is being explored by several investigators. In order to use vitamin D most efficaciously in a clinical setting, it may be beneficial to learn more about the interaction of 1,25(OH)(2)D(3) with other factors that regulate prostatic epithelial cellular growth. In this study, we examined the effect of the proliferative status of cultured cells on their ability to respond to 1,25(OH)(2)D(3), and found that minimally proliferative cells were equally as responsive to 1,25(OH)2D3 as actively dividing cells. We noted no apparent interaction of 1,25(OH)(2)D(3) with epidermal growth factor, insulin-like growth factor, cholera toxin, or transforming growth factor-β, but we did find synergistic inhibitory effects of 1,25(OH)(2)D(3) with suramin and retinoic acid. Perhaps most noteworthy was the dramatic increase in potency of 1,25(OH)(2)D(3) that occurred upon deletion of hydrocortisone from the culture medium. Our in vitro studies indicate that combination therapy of vitamin D analogs with suramin, vitamin A analogs, or anti-glucocorticoids might be considered for prostate cancer.

    View details for PubMedID 21224116

  • ALTERED GROWTH-REGULATION OF PROSTATIC EPITHELIAL-CELLS BY HUMAN PAPILLOMAVIRUS-INDUCED TRANSFORMATION INTERNATIONAL JOURNAL OF ONCOLOGY Peehl, D. M., Wong, S. T., Rhim, J. S. 1995; 6 (6): 1177-1184

    Abstract

    Six lines of human papillomavirus (HPV)-transformed prostatic epithelial cells, clonally-derived from transfection of three different parental cell strains, were analyzed for their ability to respond to stimulatory and inhibitory factors known to regulate prostatic cell growth. The cell lines were tested in a serum-free medium that was developed specifically for analysis of low-density growth of prostatic cells in order to mitigate any effects of autocrine factors. This medium has been used previously to characterize the growth regulatory pathways of primary cultures of prostatic epithelial cells derived from normal, benign prostatic hyperplasia (BPH), or adenocarcinoma tissues, as well as an SV40-transformed cell line (pRNS-1-1) that was derived from the same parental cell strain as four of the HPV-transformed lines used in the present study. The responses of the HPV-transformed cell lines to three essential mitogens in the medium - epidermal growth factor (EGF), bovine pituitary extract (BPE) and insulin-like growth factor (IGF) - were similar to those of primary cultures and pRNS-1-1, except that two of the HPV-transformed cell lines were IGF-independent for growth. The presence or absence of cholera toxin (CT), which acts synergistically with peptide growth factors, only mildly affected the proliferation of HPV-transformed cells, which is also true for primary cultures and pRNS-1-1. However, while hydrocortisone (HC) also has only minimal effect on the growth of primary cultures and pRNS-1-1, four of the HPV-transformed lines were very dependent on HC for growth. With regard to growth-inhibitory factors, all of the cell lines (HPV- or SV40- transformed) were insensitive to tumor necrosis factor-alpha (TNFalpha), which is a common feature of transformed cells. However, the cell lines remained sensitive to the growth-inhibitory properties of retinoic acid (RA) and transforming growth factor-beta (TGF(beta)). The HPV-transformed cell lines were also inhibited by 1,25(OH)(2) vitamin D-3 [1,25(OH)(2)D-3], although pRNS-1-1 cells were not. Perhaps the most unexpected finding in our study was the apparent loss of responsiveness of all of the transformed lines to fibroblast growth factor (FGF). Alterations in FGF pathways have been described for prostatic cancer cell lines and changes in expression of FGF or receptors may be involved in prostatic carcinogenesis. Our study demonstrates that the availability of primary cultures, SV40- and HPV- transformed cell lines and a well-defined culture system will provide an opportunity to characterize the processes involved in malignant transformation of prostatic cells.

    View details for Web of Science ID A1995QZ59700005

    View details for PubMedID 21556655

  • PROSTATE-SPECIFIC ANTIGEN ROLE AND FUNCTION National Conference on Prostate Cancer Peehl, D. M. WILEY-LISS. 1995: 2021–26
  • Vitamin D and prostate cancer 5th Annual Conference of the American-Institute-for-Cancer-Research on Diet and Cancer - Molecular Mechanisms of Interactions Feldman, D., Skowronski, R. J., Peehl, D. M. PLENUM PRESS DIV PLENUM PUBLISHING CORP. 1995: 53–63

    Abstract

    Our findings demonstrate the presence of VDR in various human prostate cancer cell lines and in primary cultures derived from normal, BPH and prostate cancer. In addition, 1,25-D induced several bioresponses in these cells including growth inhibition and PSA stimulation. Based on examples in many different malignant cells as well as our data in prostate cells, that vitamin D is anti-proliferative and promotes cellular maturation, it seem clear that vitamin D must be viewed as an important cellular modulator of growth and differentiation if addition to its classical role as regulator of calcium homeostasis. In this respect, vitamin D has the potential to have beneficial actions on various malignancies including prostate cancer. Its ultimate role in prostate cancer remains to be determined, but 1,25-D may prove useful in chemoprevention and/or differentiation therapy. We believe the data currently available provide the basis for an optimistic view on the possible use of vitamin D to treat prostate cancer in patients and that further investigation is clearly warranted to better define its potential therapeutic utility.

    View details for Web of Science ID A1995BD36X00005

    View details for PubMedID 7645428

  • ACTIONS OF VITAMIN-D-3 ANALOGS ON HUMAN PROSTATE-CANCER CELL-LINES - COMPARISON WITH 1,25-DIHYDROXYVITAMIN-D-3 ENDOCRINOLOGY Skowronski, R. J., Peehl, D. M., Feldman, D. 1995; 136 (1): 20-26

    Abstract

    Data from epidemiological studies has suggested that vitamin D deficiency may promote prostate cancer, although the mechanism is not understood. We have previously demonstrated the presence of vitamin D receptors (VDR) in three human prostate carcinoma cell lines (LNCaP, PC-3, and DU-145) as well as in primary cultures of stromal and epithelial cells derived from normal and malignant prostate tissues. We have also shown that 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3] can elicit an antiproliferative action in these cells. In the present study we compared the biological actions of 1,25-(OH)2D3 to those of a series of natural vitamin D3 metabolites and several synthetic analogs of vitamin D3 known to exhibit less hypercalcemic activity in vivo. In ligand binding competition experiments, we demonstrated the following order of potency in displacing [3H]1,25-(OH)2D3 from VDR: EB-1089 > 1,25-(OH)2D3 > MC-903 > 1,24,25-(OH)3D3 > 22-oxacalcitriol (OCT) > 1 alpha,25-dihydroxy-16-enecholecalciferol (Ro24-2637) > 25-hydroxyvitamin D3, with EB-1089 being approximately 2-fold more potent than the native hormone. No competitive activity was found for 25-hydroxy-16,23-diene-cholecalciferol. When compared for ability to inhibit proliferation of LNCaP cells, MC-903, EB-1089, OCT, and Ro24-2637 exhibited 4-, 3-, and 2-fold greater inhibitory activity than 1,25-(OH)2D3. Interestingly, although OCT and Ro24-2637 exhibit, respectively, 10 and 14 times lower affinity for VDR than 1,25-(OH)2D3, both compounds inhibited the proliferation of LNCaP cells with a potency greater than that of the native hormone. The relative potency of vitamin D3 metabolites and analogs to inhibit cell proliferation correlated well with the ability of these compounds to stimulate prostate-specific antigen secretion by LNCaP cells as well as with their potency to induce the 25-hydroxyvitamin D3-24-hydroxylase messenger RNA transcript in PC-3 cells. In conclusion, these results demonstrate that synthetic analogs of vitamin D3, known to exhibit reduced calcemic activity, can elicit antiproliferative effects and other biological actions in LNCaP and PC-3 cell lines. It is noteworthy that although binding to VDR is critical for 1,25-(OH)2D3 action, the analog data indicate that additional factors significantly contribute to the magnitude of the biological response. Finally, the strong antiproliferative effects of several synthetic analogs known to exhibit less calcemic activity than 1,25-(OH)2D3 suggest that these compounds potentially may be useful as an additional therapeutic option for the treatment of prostate cancer.

    View details for Web of Science ID A1995QD58400004

    View details for PubMedID 7530193

  • LIPOFECTION-MEDIATED IMMORTALIZATION OF HUMAN PROSTATIC EPITHELIAL-CELLS OF NORMAL AND MALIGNANT ORIGIN USING HUMAN PAPILLOMAVIRUS TYPE-18 DNA CANCER RESEARCH Weijerman, P. C., Konig, J. J., Wong, S. T., Niesters, H. G., Peehl, D. M. 1994; 54 (21): 5579-5583

    Abstract

    Human papillomavirus (HPV) type 18 DNA was introduced into epithelial cell strains derived from normal and cancer tissues of human prostatectomy specimens by the lipofection transfection method. Two cell lines were established: PZ-HPV-7 (transfected normal cell) and CA-HPV-10 (transfected cancer-derived cell). These lines have been maintained for over 100 passages. Incorporation of HPV type 18 DNA was confirmed by polymerase chain reaction. Immunocytochemical analysis showed expression of keratins 5 and 8, similar to the cells of origin, and the early region 6 oncoprotein of HPV. PZ-HPV-7, derived from normal diploid cells, had a modal chromosome number of 46 in early passages but became tetraploid later. CA-HPV-10 cells were aneuploid, and some retained the double minute chromosomes that were noted in the cancer-derived cells of origin. The cell lines showed a typical transformed morphology and were nontumorigenic in nude mice. We conclude that human prostatic epithelial cells derived from both normal and cancer tissues have been successfully transformed to immortality with HPV type 18 DNA. The establishment of these cell lines provides an opportunity for further development of an in vitro model of carcinogenesis for prostate cancer.

    View details for Web of Science ID A1994PP19000018

    View details for PubMedID 7923200

  • IDENTIFICATION OF INSULIN-LIKE GROWTH FACTOR-BINDING PROTEIN-3 (IGFBP-3) FRAGMENTS AND IGFBP-5 PROTEOLYTIC ACTIVITY IN HUMAN SEMINAL PLASMA - A COMPARISON OF NORMAL AND VASECTOMIZED PATIENTS JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Lee, K. O., Oh, Y. M., Giudice, L. C., Cohen, P., Peehl, D. M., Rosenfeld, R. G. 1994; 79 (5): 1367-1372

    Abstract

    Previous studies have demonstrated that insulin-like growth factor (IGF) peptides, IGF-binding proteins (IGFBPs), and IGFBP-3 proteolytic activity, are present in human seminal plasma (SP). In this study, we have further characterized the IGFBPs in SP using immunoprecipitation and Western ligand blotting, Western immunoblotting, affinity cross-linking and immunoprecipitation, and RIA of IGFBP-3 using two different assays and have identified additional proteolytic activities for IGFBP-4 and IGFBP-5 in SP. Immunoprecipitation with antibodies to IGFBP-2, IGFBP-3, and IGFBP-4, before and after affinity cross-linking, demonstrated that intact IGFBP-2 and IGFBP-4 are present in SP, but intact IGFBP-3 is absent. Low mol wt fragments of IGFBP-3, which did not bind to IGF-I or IGF-II on Western ligand blot and did not cross-link to IGF-II, were demonstrated on Western immunoblot and were measurable by two different RIAs. Proteolytic activities for IGFBP-4 and IGFBP-5 were demonstrated in SP by incubation with the respective iodinated IGFBPs. On comparing the proteolytic activity for IGFBP-4 by purified prostate-specific antigen (PSA; a known IGFBP-3 protease in SP) or by SP with measured equivalent concentrations of PSA, the dose response and fragment patterns were identical. With IGFBP-5, however, proteolysis by purified PSA was different from that by SP with measured equivalent concentrations of PSA: 1) proteolysis by pure PSA was less efficient than matched concentrations of SP; 2) the pattern of fragments after proteolysis by pure PSA was different from that after proteolysis by matched concentrations of SP; and 3) proteolysis by purified PSA was significantly inhibited by phenylmethylsulfonylfluoride and aprotinin, but proteolysis by SP was not. We conclude that human SP contains intact IGFBP-2 and IGFBP-4, but has only IGFBP-3 fragments with low affinity for IGF peptides; that PSA is able to proteolyze IGFBP-4 and IGFBP-5 (as well as IGFBP-3); and that an additional IGFBP-5 protease is probably present in SP. There was no significant difference in any of these findings in SP from normal volunteers, vasectomized patients, or patients with idiopathic azoospermia. The roles of IGFBPs and IGFBP proteases in the male reproductive system and male infertility remain to be further elucidated.

    View details for Web of Science ID A1994PT84400024

    View details for PubMedID 7525634

  • INSULIN-LIKE GROWTH-FACTOR AXIS ABNORMALITIES IN PROSTATIC STROMAL CELLS FROM PATIENTS WITH BENIGN PROSTATIC HYPERPLASIA JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Cohen, P., Peehl, D. M., Baker, B., Liu, F., Hintz, R. L., Rosenfeld, R. G. 1994; 79 (5): 1410-1415

    Abstract

    Benign prostatic hyperplasia (BPH) is a common proliferative disorder of unknown etiology. To assess whether patients with BPH have alterations in their prostatic IGF axis, we measured the expression (by Northern blotting) and the production (by Western ligand blotting and RIA) of insulin-like growth factor-II (IGF-II) and IGF-binding proteins (IGFBPs) in prostatic epithelial and stromal cell strains grown from normal (n = 7), hyperplastic (n = 7), and malignant (n = 5) surgical specimens. Levels of IGF-II messenger ribonucleic acid (mRNA; normalized for actin expression) were 10-fold higher in BPH stromal cell strains compared to those in normal stromal cell strains (P < 0.0001). Western ligand blotting of conditioned medium (CM) from normal stromal cells demonstrated the presence of IGFBP-2, -3, and -4. In the CM of BPH stromal cells, IGFBP-2 levels were dramatically reduced to less than 20% of normal (P < 0.001). Additionally, IGFBP-5, which was not observed in significant amounts in normal stromal cell-CM, was found in large quantities in BPH stromal cell-CM. Northern blot analysis of mRNA from normal and BPH stromal cells demonstrated a 5-fold decrease in IGFBP-2 mRNA (P < 0.001) and a 4-fold increase in IGFBP-5 mRNA (P < 0.01) in BPH compared to normal cells. In prostate stromal cells from cancer specimens, no abnormalities were found. No abnormalities were observed in the IGF axis parameters evaluated in prostate epithelial cells from BPH or cancer strains. We conclude that prostatic stromal cell strains isolated from patients with BPH hyperexpress the mRNA for IGF-II and IGFBP-5 while expressing reduced amounts of IGFBP-2 mRNA. IGFBP, but not IGF-II, peptide levels in CM correspond to the mRNA differences. This is the first documentation of altered gene and protein expression in this common disease. We speculate that these abnormalities in the IGF axis may be important in the pathogenesis of BPH.

    View details for Web of Science ID A1994PT84400032

    View details for PubMedID 7525636

  • BIOLOGICAL EFFECTS OF PROSTATE-SPECIFIC ANTIGEN AS AN INSULIN-LIKE GROWTH-FACTOR BINDING PROTEIN-3 PROTEASE JOURNAL OF ENDOCRINOLOGY Cohen, P., Peehl, D. M., GRAVES, H. C., Rosenfeld, R. G. 1994; 142 (3): 407-415

    Abstract

    Prostate specific antigen (PSA) is an insulin-like growth factor (IGF) binding protein-3 (IGFBP-3) protease found in seminal plasma and produced by prostatic epithelial cells (PC-E) in vivo. We examined the effects of PSA-proteolysis of IGFBP-3 on the affinity of IGFBP-3 fragments for IGFs and on the mitogenic action of IGFs on PC-E. Recombinant human IGFBP-3 was cleaved by PSA, then incubated with 125I-IGF-I or -II in the presence of varying concentrations of unlabelled peptides, and then cross-linking electrophoresis and densitometric analysis were performed. While the affinity of IGF-II for the PSA-generated IGFBP-3 fragments fell slightly compared to intact IGFBP-3, the affinity of the PSA-generated IGFBP-3 fragments for IGF-I fell by ten fold. The addition of IGF-I or -II to PC-E in serum-free culture conditions resulted in a two-fold stimulation of cell number compared to control. The presence of IGFBP-3 in the media blocked the IGF-induced stimulation, but had no independent effect in the absence of IGFs. When PSA was added to PC-E cultures to which both IGF-I or -II and IGFBP-3 were added, the inhibitory effects of IGFBP-3 on IGF mitogenesis were reversed. We conclude that PSA decreases the affinity of IGFBP-3 for IGF and can potentiate IGF action in the presence of inhibitory IGFBP-3. This phenomenon may contribute to normal and malignant prostate growth.

    View details for Web of Science ID A1994PF54800003

    View details for PubMedID 7525824

  • KERATIN EXPRESSION - A MEASURE OF PHENOTYPIC MODULATION OF HUMAN PROSTATIC EPITHELIAL-CELLS BY GROWTH-INHIBITORY FACTORS CELL AND TISSUE RESEARCH Peehl, D. M., Leung, G. K., Wong, S. T. 1994; 277 (1): 11-18

    Abstract

    Expression of certain cytokeratins can be indicative of the state of differentiation of epithelial cells. The basal cells in the normal adult human prostatic epithelium are characterized by the expression of cytokeratins 5 and 14, whereas the secretory luminal cells contain cytokeratins 8 and 18. Cells cultured from the prostatic epithelium expressed cytokeratins 5, 8, and 18, and thus had features of both basal and luminal cells. Certain growth-inhibitory conditions altered keratin expression in conjunction with growth modulation. Deletion of peptide factors and hormones from the culture medium induced the expression of cytokeratins 1 and 10, associated with a squamous phenotype. These same squamous keratins were found in very dense, stratified cultures that were maintained at confluency in standard, complete medium for extended periods. Retinoic acid enhanced the expression of secretory luminal cell-associated cytokeratins 8 and 18 in semi-confluent cultures. Other growth inhibitory factors such as suramin, transforming growth factor-beta, and interferon-gamma had no effect on keratin expression. These observations indicate that the differentiation of prostatic epithelial cells can be directed toward alternate pathways, either squamous or secretory, by different growth-inhibitory conditions. However, not all growth inhibitory factors altered differentiation, demonstrating that growth inhibition in itself is not a sufficient inducer of differentiation.

    View details for Web of Science ID A1994NU28100002

    View details for PubMedID 7519968

  • CHARACTERIZATION OF ADULT HUMAN PROSTATIC EPITHELIAL-CELLS IMMORTALIZED BY POLYBRENE-INDUCED DNA TRANSFECTION WITH A PLASMID CONTAINING AN ORIGIN-DEFECTIVE SV40-GENOME INTERNATIONAL JOURNAL OF ONCOLOGY Lee, M. S., GARKOVENKO, E., Yun, J. S., Weijerman, P. C., Peehl, D. M., Chen, L. S., Rhim, J. S. 1994; 4 (4): 821-830

    Abstract

    Normal adult human prostatic epithelial cells were infected with an adenovirus 12-SV40 virus or transfected by polybrene-induced gene transfer with a plasmid (pRSV-T) containing the SV40 early region genes or with a plasmid (pRNS-1) containing an origin-defective SV40 genome and a plasmid carrying the neomycin resistance gene. Colonies of morphologically altered cells were isolated, cultured in a serum-free medium and characterized. These cells had extended lifespan in culture compared to normal adult human prostatic epithelial cells. Both Ad12-SV40-infected and pRSV-T-transfected cultures eventually underwent senescence. pRNS-1-transfected cells (pRNS-1-1), however, have now been grown for more than 50 passages. These cells contain the SV40 genome, express SV40 T-antigen, and are not tumorigenic in nude mice. They express cytokeratins 5 and 8, like the parent cells, and are pseudodiploid. Analysis of growth regulatory processes revealed that the growth of pRNS-1-1 cells was stimulated similarly to that of normal prostatic epithelial cells by epidermal growth factor, insulin-like growth factor, and pituitary extract. The response of pRNS-1 cells to a growth-inhibitory factor, retinoic acid, was also similar to that of normal cells. However, pRNS-1-1 cells were less responsive than normal cells to growth inhibition by transforming growth factor-beta, and had lost altogether the ability of normal cells to be inhibited by tumor necrosis factor-alpha and 1,25 (OH)2 vitamin D3. Therefore transformation appeared to alter growth-inhibitory but not growth - stimulatory mechanisms. These cells should be useful in elucidating the multistep mechanism of carcinogenesis of the prostate.

    View details for Web of Science ID A1994ND85600006

    View details for PubMedID 21566988

  • ANTIPROLIFERATIVE EFFECTS OF 1,25-DIHYDROXYVITAMIN-D(3) ON PRIMARY CULTURES OF HUMAN PROSTATIC CELLS CANCER RESEARCH Peehl, D. M., Skowronski, R. J., Leung, G. K., Wong, S. T., Stamey, T. A., Feldman, D. 1994; 54 (3): 805-810

    Abstract

    Cultures of adult human prostatic epithelial and fibroblastic cells were established from normal, benign hyperplastic, and malignant tissues. Vitamin D receptors were detected by ligand binding of [3H]1,25-dihydroxyvitamin D3 [1,25(OH)2D3] in cytosolic extracts prepared from all types of cell cultures as well as from fresh prostatic tissues. Vitamin D receptor transcripts were demonstrated by Northern blot analysis. 1,25-(OH)2D3 inhibited the growth of epithelial cells with half-maximal inhibition at approximately 1 nM. The growth of fibroblasts was also inhibited by 1,25(OH)2D3 but to a lesser extent. This is consistent with the apparently lower level of vitamin D receptors in fibroblasts compared to epithelial cells determined by ligand binding and Northern analysis of RNA transcripts. The growth inhibition of epithelial cells by 1,25(OH)2D3 was irreversible even after a short 2-h exposure, but morphology and keratin expression were not appreciably altered by long-term exposure to the hormone. A physiological role for 1,25(OH)2D3 in the prostate is postulated, and the inhibitory effect of 1,25(OH)2D3 on cancer-derived prostate cells may provide a basis for new preventive or therapeutic strategies.

    View details for Web of Science ID A1994MU67700036

    View details for PubMedID 7508338

  • THE IGF AXIS IN THE PROSTATE HORMONE AND METABOLIC RESEARCH Cohen, P., Peehl, D. M., Rosenfeld, R. G. 1994; 26 (2): 81-84

    Abstract

    The insulin-like growth factor (IGF) axis is a multi-component network of molecules involved in the regulation of cell growth. The axis includes two major ligands, (IGF-I and IGF-II), cell surface receptors, (the type I IGF receptor family as well as the type II IGF receptor), a family of high affinity binding proteins which regulate IGF availability to the receptors, (the IGFBPs), and a group of IGFBP proteases which cleave IGFBBPs and modulate IGF action. Human seminal plasma contains IGF-I and -II, IGFBP-2 and -4, as well as IGFBP-3 fragments and IGFBP-3 protease activity. A prostatic source for these IGF-axis molecules is likely. We have demonstrated the human prostate to contain all the elements of a functional IGF system. Prostate fibroblasts in primary culture (PC-F) express mRNA for IGF-II and produce IGF-II peptide in biologically active concentrations. This IGF-II appears to be of a high molecular weight (15 kD) species. Prostate epithelial cells in primary culture (PC-E) express the type I IGF receptor. These cells also produce IGFBP-2 and -4, (on both mRNA and peptide levels), while PC-F secrete IGFBP-2, -3 and -4. PC-E are exquisitely sensitive to the mitogenic effects of IGFs. Finally, prostate specific antigen (PSA), secreted from PC- and found in seminal plasma, can function as a potent IGFBP-3 protease. This IGFBP-3 protease activity can remove the inhibitory effects of IGFBP-3 on IGF-I induced PC-3 growth.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1994MY29500001

    View details for PubMedID 8200618

  • Human papilloma virus type 18 DNA immortalized cell lines from the human prostate epithelium. Progress in clinical and biological research Weijerman, P. C., ROMIJN, H. C., Peehl, D. M. 1994; 386: 67-69

    View details for PubMedID 7800701

  • THE IGF AXIS IN PROSTATIC DISEASE 3rd International Symposium on Insulin-Like Growth Factors Cohen, P., Peehl, D. M., Bhala, A., Dong, G., Hintz, R. L., Rosenfeld, R. G. ELSEVIER SCIENCE PUBL B V. 1994: 369–377
  • INVITRO PROSTATE CANCER DRUG SCREEN 5th Tokyo Symposium on Prostate Cancer - Fundamental Approaches to the Diagnosis and Treatment for Prostate Cancer and BPH Peehl, D. M., Erickson, E., Malspeis, L., Mayo, J., Camalier, R. F., Monks, A., CRONISE, P., Paull, K., Grever, M. R. ADENINE PRESS. 1994: 203–207
  • HUMAN PAPILLOMA-VIRUS TYPE-18 DNA IMMORTALIZED CELL, LINES FROM THE HUMAN PROSTATE EPITHELIUM 2nd International Congress of the Dutch-Urological-Association: New Insights into the Aetiology, Diagnosis and Management of BPH Weijerman, P. C., ROMIJN, H. C., Peehl, D. M. WILEY-LISS, INC. 1994: 67–69
  • Actions of 1,25-dihydroxyvitamin D and synthetic analogs on cultured human prostate carcinoma cells 9th Workshop on Vitamin D Skowronski, R. J., Peehl, D. M., Cramer, S., Feldman, D. WALTER DE GRUYTER GMBH. 1994: 520–521
  • VITAMIN-D AND PROSTATE CANCER 5th Tokyo Symposium on Prostate Cancer - Fundamental Approaches to the Diagnosis and Treatment for Prostate Cancer and BPH Peehl, D. M., Skowronski, R. J., Feldman, D. ADENINE PRESS. 1994: 57–61
  • ROLE OF VITAMIN-D-RECEPTORS IN PROSTATE CANCER International Symposium on Sex Hormones and Antihormones in Endocrine Dependent Pathology - Basic and Clinical Aspects Peehl, D. M., Skowronski, R. J., Feldman, D. ELSEVIER SCIENCE PUBL B V. 1994: 23–31
  • PR1 - A MONOCLONAL-ANTIBODY THAT REACTS WITH AN ANTIGEN ON THE SURFACE OF NORMAL AND MALIGNANT PROSTATE CELLS JOURNAL OF THE NATIONAL CANCER INSTITUTE PASTAN, LOVELACE, E., RUTHERFORD, A. V., KUNWAR, S., WILLINGHAM, M. C., PEEHL, D. M. 1993; 85 (14): 1149–54

    Abstract

    The principal treatment for prostate cancer is surgery; prostate cancer is resistant to the common anticancer drugs. The only useful therapy for metastases involves diminishing testosterone levels by orchiectomy or administration of drugs, either of which may increase survival time. One approach to prostate cancer treatment is to use a monoclonal antibody (MAb) to target cytotoxic substances to these cancer cells. The MAbs available either do not react uniformly with prostate cancer cells or react with normal tissues. Thus, a new MAb is needed.The goal of this study was to isolate an MAb that reacts with an antigen present on the surface of prostate cancer cells.A strain of prostate cancer cells was isolated from a prostate cancer specimen, grown for 2-4 weeks in short-term culture, and used to immunize BALB/c mice. Hybridomas were then prepared by using spleen cells from the immunized mice. One hybridoma produced an MAb (PR1) that reacted with prostate cancers.MAb PR1 is an IgMK subtype that reacts uniformly with the surface of most (25 of 26) adenocarcinomas of the prostate. It also reacts with the surface antigen on normal prostate epithelial cells and on cells from benign prostatic hyperplasia. MAb PR1 reacts with a limited number of normal tissues including a subset of principal cells located in the collecting ducts of the kidney.We conclude that MAb PR1 reacts with a differentiation antigen present in normal prostate and that this antigen continues to be expressed on almost all adenocarcinomas of the prostate.This antibody may be useful for the diagnosis of or therapy for prostate cancer.

    View details for DOI 10.1093/jnci/85.14.1149

    View details for Web of Science ID A1993LM44900014

    View details for PubMedID 7686582

  • VITAMIN-D AND PROSTATE-CANCER - 1,25-DIHYDROXYVITAMIN-D3 RECEPTORS AND ACTIONS IN HUMAN PROSTATE-CANCER CELL-LINES ENDOCRINOLOGY Skowronski, R. J., Peehl, D. M., Feldman, D. 1993; 132 (5): 1952-1960

    Abstract

    It has been suggested that vitamin D deficiency may promote prostate cancer, although the mechanism is not understood. In this study three human prostate carcinoma cell lines, LNCaP, DU-145, and PC-3, were examined both for the presence of specific 1,25 dihydroxyvitamin D3 [1,25(OH)2D3] receptors (VDRs) and also employed to study the effects of hormone on cell proliferation and differentiation. Ligand binding experiments demonstrated classical VDR in all three cell lines examined with an apparent dissociation constant of 7.5, 5.4, and 6.3 x 10(-11) M for LNCaP, DU-145, and PC-3 cells, respectively. Corresponding binding capacity for the three prostate carcinoma cell lines were 27, 31, and 78 fmol/mg protein, respectively. The presence of VDR in the three cell lines was also confirmed by immunocytochemistry. In addition, one major 4.6-kilobase messenger RNA transcript hybridizing with a specific human VDR complementary DNA probe was identified in all three cell lines. Interestingly, both DU-145 and PC-3 but not LNCaP cell lines exhibited 1,25(OH)2D3-stimulated induction of 24-hydroxylase messenger RNA employed as a marker of 1,25(OH)2D3 action. Physiological levels of 1,25(OH)2D3 dramatically inhibited proliferation of the LNCaP and PC-3 cell lines. However, in spite of the presence of high affinity VDR, proliferation of DU-145 cells was not inhibited by 1,25(OH)2D3 at the doses tested. Treatment with 1,25(OH)2D3 caused a dose-dependent stimulation of prostate-specific antigen secretion by LNCaP cells. In conclusion, these results demonstrate that these three human prostate carcinoma cell lines all possess specific VDR and that 1,25(OH)2D3 treatment can elicit both an antiproliferative and a differentiating action on these cancer cells. The findings lend support to the hypothesis that vitamin D might exert beneficial actions on prostate cancer risk.

    View details for Web of Science ID A1993KZ69900011

    View details for PubMedID 7682937

  • ELEVATED LEVELS OF INSULIN-LIKE GROWTH FACTOR-BINDING PROTEIN-2 IN THE SERUM OF PROSTATE-CANCER PATIENTS JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Cohen, P., Peehl, D. M., Stamey, T. A., Wilson, K. F., Clemmons, D. R., Rosenfeld, R. G. 1993; 76 (4): 1031-1035

    Abstract

    We have previously documented the presence of specific insulin-like growth factor (IGF)-binding protein (IGFBPs) in seminal plasma and prostate epithelial cell-conditioned medium IGFBP-2 is the prevalent IGFBP in both fluids. To assess whether patients with prostate carcinoma have alterations in serum IGFP levels related to the production of IGFBPs by their tumors, we performed Western ligand blots (WLB) and IGFBP-2 RIA on serum samples from 32 patients with prostate carcinoma of various degrees of clinical severity and compared them to results in 16 healthy age-matched controls. We have also measured serum IGF-I and -II by RIA. The mean level of IGFBP-2 in the prostate cancer patients was 170% of control levels by WLB analysis and 195% of control levels by RIA (P < 0.01). The degree of elevation of IGFBP-2 was related to the stage of the tumor and the levels of the serum tumor marker, prostate-specific antigen. Serum IGFBP-3 levels determined by WLB and serum IGF-I and IGF-II levels measured by RIA after acid chromatography were not different among the subjects with cancer and the normal controls. We conclude that IGFBP-2, which is the main IGFBP produced by prostate epithelial cells, is elevated in the serum of patients with prostate carcinoma, and that the degree of this elevation is related to serum prostate-specific antigen levels and the stage of the tumor. We speculate that prostate-derived IGFBPs may be secreted by prostate tumors and could e of value in understanding the pathophysiology of prostatic tumor growth as well as provide potential diagnostic markers.

    View details for Web of Science ID A1993KW77300041

    View details for PubMedID 7682560

  • ONCOGENES IN PROSTATE-CANCER - AN UPDATE 1ST NATIONAL CONF ON PROSTATE CANCER Peehl, D. M. WILEY-LISS. 1993: 1159–64

    Abstract

    Oncogenes have been implicated in the carcinogenic development of many diverse types of human malignancies. For some cancers, the expression of specific oncogenes has been shown to have diagnostic or prognostic value. By contrast currently, no oncogene has been correlated conclusively with the initiation or progression of prostate cancer. The ras oncogene has been investigated the most thoroughly for its involvement in prostate cancer, but ras does not appear to play a significant role in the development of this malignancy. Several years ago, limited studies hinted at the possibility of overexpression of the myc oncogene and aberrant expression of the sis oncogene in prostate cancer, but additional studies to clarify the involvement of these oncogenes have not been done. Oncogenic activity of growth factors or growth factor receptors in prostate cancer has been suggested but not amply demonstrated. Current dogma indicates that oncogenes exist in prostate cancer, but these will be identified only by more intensive investigation.

    View details for Web of Science ID A1993KM67400038

    View details for PubMedID 8428339

  • VITAMIN-D AND PROSTATE-CANCER - 1,25-DIHYDROXYVITAMIN D3 RECEPTORS AND ACTIONS IN HUMAN PROSTATE-CANCER CELL-LINES SKOWRONSKI, R. J., PEEHL, D. M., FELDMAN, D. SLACK INC. 1993: A45
  • VITAMIN-A REGULATES PROLIFERATION AND DIFFERENTIATION OF HUMAN PROSTATIC EPITHELIAL-CELLS PROSTATE Peehl, D. M., Wong, S. T., Stamey, T. A. 1993; 23 (1): 69-78

    Abstract

    The response of cultured human prostatic epithelial cells to vitamin A was measured by clonal growth assay in serum-free medium. Retinoic acid at 3 nM or higher inhibited the proliferation of cell strains derived from normal, benign hyperplastic and malignant tissues, while lower levels (0.03 nM) were stimulatory. Reduced proliferation induced by retinoic acid was accompanied by a marked change in morphology, as intercellular adhesion decreased. In conjunction, the expression of keratins 8 and 18, associated with the differentiated luminal phenotype of prostatic epithelia, was increased. In post-confluent cultures, retinoic acid prevented the appearance of keratin 1, which accompanied the development of a squamous phenotype by cells maintained under these conditions. The findings of this study indicate a role for vitamin A as a modulator of the growth and differentiation of prostatic epithelial cells.

    View details for PubMedID 7687781

  • PROSTATE-SPECIFIC ANTIGEN (PSA) IS AN INSULIN-LIKE GROWTH-FACTOR BINDING PROTEIN-3 PROTEASE FOUND IN SEMINAL PLASMA JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Cohen, P., GRAVES, H. C., Peehl, D. M., Kamarei, M., Giudice, L. C., Rosenfeld, R. G. 1992; 75 (4): 1046-1053

    Abstract

    Insulin-like growth factor binding protein-3 (IGFBP-3), the major serum carrier protein for the IGFs, is absent from Western ligand blots of seminal plasma, but is detectable by RIA. IGFBP-3 protease activity has recently been described in pregnancy serum. We investigated the possibility that seminal plasma contains an IGFBP-3 protease, by incubating seminal plasma with 125I-labeled human IGFBP-3. Seminal plasma was found to have potent IGFBP-3 protease activity with a cleavage pattern different from that of pregnancy serum. Prostate-specific antigen (PSA) is a serine protease found in semen. Autoradiographs measuring IGFBP-3 protease activity demonstrated that purified PSA cleaved IGFBP-3, yielding a cleavage pattern identical to that of seminal plasma. IGFBP-2 and -4 in seminal plasma were not degraded by PSA. Cleavage of IGFBP-3 by PSA resulted in a marked reduction in the binding affinity of the fragments to IGF-I, but not IGF-II. We speculate that PSA may serve to modulate IGF function within the reproductive system or in prostate cancer by altering IGF-IGFBP-3 interactions.

    View details for Web of Science ID A1992JT81500015

    View details for PubMedID 1383255

  • ANALYSIS OF PROSTATIC TUMOR CULTURES USING FLUORESCENCE INSITU HYBRIDIZATION (FISH) CANCER GENETICS AND CYTOGENETICS Brothman, A. R., Patel, A. M., Peehl, D. M., Schellhammer, P. F. 1992; 62 (2): 180-185

    Abstract

    Analysis of ten primary prostatic tumor cultures using fluorescence in-situ hybridization (FISH) with pericentromeric probes for chromosomes 7, 8, 10, 16, 17, and 18 revealed aneusomies in nine of these specimens. Classical cytogenetics by G-banding indicated that only four of those same ten specimens had any (but not consistent) clonal abnormalities. This preliminary study suggests that aneusomy is a common event in early-stage prostatic tumors, and also supports the notion that multiple chromosomes are involved. In combination with routine cytogenetic analysis, FISH is thus likely to be a powerful tool in the evaluation of prostatic cancer.

    View details for Web of Science ID A1992JW68400012

    View details for PubMedID 1394106

  • INSULIN-LIKE GROWTH-FACTORS (IGFS), IGF RECEPTORS, AND IGF-BINDING PROTEINS IN PRIMARY CULTURES OF PROSTATE EPITHELIAL-CELLS JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Cohen, P., Peehl, D. M., Lamson, G., Rosenfeld, R. G. 1991; 73 (2): 401-407

    Abstract

    Insulin-like growth factors (IGFs) are potent mitogens that bind with high affinity and specificity to IGF receptors and IGF-binding proteins (IGFBPs). We studied the roles of these three groups of proteins in prostate epithelial cells (PEC) in primary culture grown under serum-free conditions. Affinity cross-linking of IGF-I and IGF-II to crude membranes prepared from PEC revealed an abundance of type 1 IGF receptors and no evidence of type 2 IGF receptors. Western ligand blots of conditioned media (CM) from PEC demonstrated the presence of two specific IGFBP bands similar to those previously demonstrated in seminal plasma, with approximate mol wt of 31 and 24 kDa. The 31-kDa band was immunoprecipitable with an antibody to IGFBP-2, and neither band could be deglycosylated with endoglycosidase-F. Northern blot analysis of poly(A)+ RNA prepared from PEC with cDNAs for hIGFBP-1, -2, and -3 documented the expression of mRNA for hIGFBP-2 only. Modifications of the serum-free conditions of PEC did not significantly alter the IGFBP profile of PEC CM. The ability of IGF-I, IGF-II, and insulin to stimulate clonal growth of PEC was examined. IGF-I stimulated PEC growth with an ED50 of 0.1 ng/mL. IGF-II and insulin, respectively, were 1 and 3 orders of magnitude less effective than IGF-I in stimulating the growth of PEC. Radioimmunoassayable IGF-I and IGF-II levels in PEC CM were below the assay detection levels. In conclusion, we suggest that IGFs are important growth stimulators of PEC in culture, that their actions are mediated through the type 1 IGF receptor, and that PEC produce hIGFBP-2 and a 24-kDa IGFBP which may modulate IGF action in these cells.

    View details for Web of Science ID A1991FY51100029

    View details for PubMedID 1713219

  • CYTOGENETIC EVALUATION OF 20 CULTURED PRIMARY PROSTATIC TUMORS CANCER GENETICS AND CYTOGENETICS Brothman, A. R., Peehl, D. M., Patel, A. M., MACDONALD, G. R., McNeal, J. E., LADAGA, L. E., Schellhammer, P. F. 1991; 55 (1): 79-84

    Abstract

    We report the cytogenetic evaluation of 20 cultured cell strains derived from primary prostatic adenocarcinomas obtained from radical prostatectomies. The majority of the strains contained cells with only normal male karyotypes (46,XY), but cytogenetically abnormal clonal populations were found in five strains. Two of those strains contained aberrations involving the Y chromosome, one with a -Y and one with a +Y. Three strains (one of which also had the XYY karyotype) exhibited cells with double-minute chromosomes and four strains contained near tetraploid cells.

    View details for Web of Science ID A1991GH73400011

    View details for PubMedID 1913611

  • INVITRO PROTON SPECTROSCOPY OF NORMAL AND ABNORMAL PROSTATE MAGNETIC RESONANCE IN MEDICINE YACOE, M. E., Sommer, G., Peehl, D. 1991; 19 (2): 429-438

    Abstract

    Previous biochemical and 13C NMR spectroscopic data have suggested that the metabolism of citrate, a secretory product of normal prostate, may be interrupted in prostate cancer. In the present study in vitro 1H NMR spectroscopy was used to see if cell strains derived from prostate cancers could be reliably distinguished from those of normal prostate epithelium. High-resolution one-dimensional and two-dimensional J-resolved 1H NMR spectra as well as gas chromatography coupled with mass spectroscopy were used to study extracts of highly defined cell strains from normal peripheral zone, normal central zone, adenocarcinoma, and benign prostatic hyperplasia. Resonances assigned to citric acid and related metabolites were identified. Cell strains derived from prostate cancers tended to have smaller amounts of citrate than those from normal prostate epithelium. However, the differences were small and not statistically significant. The lack of statistically significant differences may reflect the variability present in both normal and abnormal cell strains and thus underscore the well-known difficulty in differentiating normal and cancerous tissues.

    View details for Web of Science ID A1991FQ24300033

    View details for PubMedID 1715505

  • CYTOSTATIC EFFECTS OF SURAMIN ON PROSTATE-CANCER CELLS CULTURED FROM PRIMARY TUMORS JOURNAL OF UROLOGY Peehl, D. M., Wong, S. T., Stamey, T. A. 1991; 145 (3): 624-630

    Abstract

    Suramin is currently undergoing clinical trials as a chemotherapeutic agent for prostate cancer. The effects of suramin on cultured human epithelial cells derived from normal, benign hyperplastic, and malignant prostate tissues were examined. In serum-free medium, suramin inhibited the clonal growth of prostate cells at a half-maximal dose of approximately 10 micrograms/ml. Growth inhibition by suramin was completely reversible even after 24 hours of exposure. In conjunction, suramin did not alter cellular phenotype with regard to expression of keratins and prostate-specific antigens. Although suramin is reportedly an antagonist of growth factor-mediated mitogenesis, ten-fold excesses of growth factors did not appreciably suppress the cytostatic activity of suramin. In comparison to the activities of other possible chemotherapeutic agents, suramin would appear suboptimal because its inhibitory effects are reversible and it does not induce a terminally differentiated cellular phenotype.

    View details for Web of Science ID A1991FA69900038

    View details for PubMedID 1705296

  • CULTURE OF PROSTATIC EPITHELIAL-CELLS FROM ULTRASOUND-GUIDED NEEDLE BIOPSIES PROSTATE Peehl, D. M., Wong, S. T., Terris, M. K., Stamey, T. A. 1991; 19 (2): 141-147

    Abstract

    A protocol which was developed for the culture of epithelial cells from radical prostatectomy specimens was slightly modified to permit the culture of cells from ultrasound-guided prostatic needle biopsies. The collagenase digestion step of the standard protocol was omitted, and biopsies were simply minced and allowed to attach to collagen-coated dishes in serum-free medium. Cell outgrowths from biopsies were free of fibroblasts, and expression of keratin, prostate specific antigen, and prostatic acid phosphatase was maintained in vitro. The establishment of a bank of frozen cells from primary cultures permitted repetitive studies with individual cell strains, which could be serially passaged and were capable of clonal growth. The ability to derive cultures from biopsies will facilitate the biological characterization of cells from primary prostate tumors of high malignant grade, which are not commonly available from radical prostatectomy specimens.

    View details for Web of Science ID A1991GW40600006

    View details for PubMedID 1717964

  • SEARCHING FOR SUPPRESSOR GENES IN PROSTATE-CANCER CANCER SURVEYS Peehl, D. M. 1991; 11: 25-34

    Abstract

    Prostate cancer poses many challenges for clinicians. Methods for early detection are not satisfactory, and in fact more efficient detection will lead to even further dilemmas. We know from necropsy studies that the occurrence of prostate cancer is much higher than the clinical incidence (Yatani et al, 1982). Obviously, only some of the prostate cancers detected early would progress to a health threatening state if left untreated--the question is, which ones? This is a question that must be addressed, because prostate cancer has the highest occurrence rate of any cancer among males in the US, and current approaches to eradicating early stage prostate cancers involve expensive surgical or irradiation techniques. Even for primary prostatic tumours of more advanced stages, current prognostic indicators are not ideal. What is the potential of specific suppressor genes lost in prostate cancer for identifying useful prognostic indicators? Even though numerous molecular events, including loss as well as gain of gene activity, have been delineated for colon cancer, no single genetic change has proven prognostic. Rather, increasing malignant potential of colon cancer seems to be correlated with an accumulation of several genetic events. On the other hand, a specific chromosomal abnormality, a deletion of chromosome 17p, does appear to be a prognostic indicator for distinguishing high grade from low grade transitional cell carcinoma (Olumi et al, 1990). Whether loss of chromosome 17p will prove to be a better prognostic marker than other parameters, however, is still unknown. At present, no consistent genetic changes have been associated with any stage of prostate cancer, so we are a long way from determining whether such changes will prove their hypothetical usefulness as new and improved prognostic indicators. The other clinical realm in which the identification of loss of suppressor genes in prostate cancer might prove a resource is therapy. Suppressor gene products obviously provide key regulatory functions, perhaps linked to differentiation, in normal cells. Restoration of these regulatory functions in cancer cells, either by directly supplying the lost regulatory element or by circumventing the missing element by other means, is a novel therapeutic approach that might offer hope even in metastatic disease. Realization of this dream will require much more knowledge of the precise nature of the suppressor gene products lost in cancer, and a detailed understanding of the hierarchy of regulation of growth and differentiation in normal cells.(ABSTRACT TRUNCATED AT 400 WORDS)

    View details for Web of Science ID A1991HQ11500004

    View details for PubMedID 1668728

  • DOWN-REGULATION OF A CALMODULIN-RELATED GENE DURING TRANSFORMATION OF HUMAN MAMMARY EPITHELIAL-CELLS PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Yaswen, P., SMOLL, A., Peehl, D. M., Trask, D. K., Sager, R., Stampfer, M. R. 1990; 87 (19): 7360-7364

    Abstract

    A human cDNA library obtained from cultured normal mammary epithelial cells (HMECs) was searched by subtractive hybridization for genes whose decrease in expression might be relevant to epithelial transformation. One clone identified by this procedure corresponded to a 1.4-kilobase mRNA, designated NB-1, whose expression was decreased greater than 50-fold in HMECs tumorigenically transformed in vitro after exposure to benzo[a]pyrene and Kirsten sarcoma virus. Sequence analysis of NB-1 cDNA revealed an open reading frame with a high degree of homology to calmodulin. NB-1 expression could be demonstrated by polymerase chain reaction amplification in normal breast, prostate, cervix, and epidermal tissues. The presence of NB-1 transcripts was variable in primary breast carcinoma tissues and undetectable in tumor-derived cell lines of breast, prostate, or other origins. NB-1 mRNA expression could be down-regulated in cultured HMECs by exposure to reconstituted extracellular matrix material, while exposure to transforming growth factor type beta increased its relative abundance. The protein encoded by NB-1 may have Ca2+ binding properties and perform functions similar to those of authentic calmodulin. Its possible roles in differentiation and/or suppression of tumorigenicity in epithelial tissues remain to be examined.

    View details for Web of Science ID A1990EB29900004

    View details for PubMedID 2217169

  • FREQUENCY AND PATTERN OF KARYOTYPIC ABNORMALITIES IN HUMAN PROSTATE-CANCER CANCER RESEARCH Brothman, A. R., Peehl, D. M., Patel, A. M., McNeal, J. E. 1990; 50 (12): 3795-3803

    Abstract

    The cytogenetic evaluation of 30 cultured primary prostatic cancer specimens obtained during radical prostatectomies of patients with relatively early stage disease is reported. The majority of specimens examined showed a normal male karyotype, 46,XY. Nine samples contained clonally abnormal populations including five specimens which were hyperdiploid (modal range, 65-92 chromosomes), one specimen containing double minute chromosomes, and three containing structural aberrations. Loss of the Y chromosome and a partial trisomy for chromosome 4 was observed in a sample from one patient. Another sample showed a translocation between the long arms of chromosomes 5 and 7. The only tumor obtained from a previously irradiated patient contained no normal cells, a modal chromosome number of 45, loss of chromosomes 2 and Y, and multiple structural rearrangements. The appearance of any clonal cytogenetic abnormality correlated in general with a poorly differentiated state of cancer. A survey of all available previous cytogenetic data on human prostate adenocarcinoma indicated that the loss of chromosomes 1, 2, 5, and Y, the gain of chromosomes 7, 14, 20, and 22, and rearrangements involving chromosome arms 2p, 7q, and 10q are the most common changes observed. This suggests that, although the assignment of a single chromosomal aberration as a marker for early stage prostatic cancer is unlikely, several consistent "hotspots" might be of significance in the etiology of this disease.

    View details for Web of Science ID A1990DG94700052

    View details for PubMedID 2340524

  • ANALYSIS OF SOMATIC-CELL HYBRIDS DERIVED FROM NORMAL HUMAN PROSTATIC EPITHELIAL-CELLS FUSED WITH HELA-CELLS PROSTATE Peehl, D. M., Wong, S. T., McNeal, J. E., Stamey, T. A. 1990; 17 (2): 123-136

    Abstract

    Somatic cell hybrids have been instrumental in the recognition of specific chromosomes containing genes capable of suppressing the malignant phenotype. As a first step towards the identification of possible suppressor genes in prostate cells, we created hybrids by fusing normal prostate cells with malignant HeLa cells. Similar to hybrids made with other combinations of normal and malignant cells, the normal phenotype was dominant and the malignant phenotype was suppressed. The phenotype of the nontumorigenic hybrids after injection into nude mice resembled that of normal keratinocyte X HeLa hybrids, and tiny, nonprogressive keratinized nodules were produced. One hybrid clone was tumorigenic, possible due to the loss of a normal suppressor gene, and displayed glandular as well as squamous elements. Further characterization of these hybrids should permit isolation of specific suppressor genes, as well as promote recognition of elements that regulate the glandular phenotype.

    View details for Web of Science ID A1990DZ96800005

    View details for PubMedID 1697966

  • TRANSFORMATION OF HUMAN NEONATAL PROSTATE EPITHELIAL-CELLS BY STRONTIUM PHOSPHATE TRANSFECTION WITH A PLASMID CONTAINING SV40 EARLY REGION GENES CANCER RESEARCH Kaighn, M. E., Reddel, R. R., Lechner, J. F., Peehl, D. M., Camalier, R. F., Brash, D. E., Saffiotti, U., Harris, C. C. 1989; 49 (11): 3050-3056

    Abstract

    Neonatal human prostatic epithelial cells (NP-2s) were transfected by strontium phosphate coprecipitation with a plasmid (pRSV-T) containing the SV40 early region genes. The cells transfected with pRSV-T, but not the sham-transfected controls, formed rapidly growing, multilayered colonies within 2 weeks at a frequency of 1 x 10(-4) in a serum-free medium (P4-8F). In all, 28 colonies of transformed cells were isolated. Three of these have been cultured for a sufficient length of time to show that their growth potentials are well beyond that of the normal progenitor cells (NP-2s). There is also little or no indication of the culture "crisis" commonly seen in SV40-transformed cells in these transfected lines. All contain cytokeratins and SV40 T-antigen as revealed by immunofluorescence, have ultrastructural features of epithelial cells, and are pseudodiploid. None have produced tumors within 1 year after s.c. injection into nude mice. The transformed as well as the parental NP-2s cells require bovine pituitary extract for growth in serum-free medium and are stimulated by transforming growth factor beta 1 (TGF-beta 1) and epidermal growth factor in clonal growth assays. In contrast, a prostatic carcinoma cell line (PC-3) is inhibited by TGF-beta 1. This serum-free system and immortalized transfected clones will be useful for studying the action of putative prostatic carcinogens and tumor-promoting agents.

    View details for Web of Science ID A1989U695200040

    View details for PubMedID 2541897

  • CLONAL GROWTH OF HUMAN PROSTATIC EPITHELIAL-CELLS IS STIMULATED BY FIBROBLASTS PROSTATE Kabalin, J. N., Peehl, D. M., Stamey, T. A. 1989; 14 (3): 251-263

    Abstract

    The interaction of fibroblasts with adult human prostatic epithelial cells was studied in vitro. Stimulation of epithelial cell proliferation, measured by clonal growth assay, was demonstrated when prostatic epithelial cells were grown in coculture with fibroblasts. Epithelial growth in cocultures with fibroblasts was greater than could be obtained in isolated culture in an "optimized," serum-free medium previously described. Fibroblasts were able to compensate for the deletion of several growth factors from this "optimized" medium, including epidermal growth factor and insulin, but were notably unable to replace bovine pituitary extract. Epithelial growth stimulation identical to that achieved in coculture was produced by medium conditioned by fibroblasts. Similar results were obtained by using fibroblasts of adult human prostate origin, adult human skin, human fetal lung fibroblasts (IMR-90), and mouse 3T3 cells. No difference in response was demonstrated between prostatic epithelial cells derived from normal or malignant tissues. Fixed fibroblast monolayers and extracellular matrix prepared from fibroblast cultures failed to stimulate prostatic epithelial growth. These results suggest that a soluble growth factor is secreted by prostatic fibroblasts and other human fibroblasts of nonprostatic origin, as well as by embryonic mesenchymal cells from nonhuman species, which is capable of producing a marked proliferative response in vitro from adult human prostatic epithelium. This proliferative response could not be reproduced by the addition of a variety of known growth factors to prostatic epithelial cell cultures.

    View details for Web of Science ID A1989AA83000005

    View details for PubMedID 2499876

  • In vitro studies of human prostatic epithelial cells: attempts to identify distinguishing features of malignant cells. Growth factors Peehl, D. M., Wong, S. T., Bazinet, M., Stamey, T. A. 1989; 1 (3): 237-250

    Abstract

    Recent advances in culture techniques have enabled routine establishment and propagation of epithelial cells derived from normal and malignant tissues of the human prostate. Comparative studies of the responses of normal and cancer-derived cell populations to various growth and differentiation factors in vitro were undertaken to examine the possibility that cancer cells might respond differentially. Clonal growth assays in serum-free medium demonstrated that optimal proliferation of normal as well as cancer cell strains was generally dependent on the presence of cholera toxin, epidermal growth factor, pituitary extract, hydrocortisone, insulin, and high levels of calcium in the culture medium, and on the use of collagen-coated dishes. Only one cancer strain responded aberrantly to epidermal growth factor and hydrocortisone. Putative differentiation factors (transforming growth factor-beta and vitamin A) inhibited the growth of all normal and cancer strains. The origin of a cancer-derived cell strain that responded similarly to normal strains was verified by positive labeling with a prostate cancer-specific antibody, validating the conclusion from these studies that normal and cancer prostatic epithelial cells are not distinguishable on the basis of responses to the tested factors.

    View details for PubMedID 2698220

  • CLONAL GROWTH-CHARACTERISTICS OF ADULT HUMAN PROSTATIC EPITHELIAL-CELLS IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY Peehl, D. M., Wong, S. T., Stamey, T. A. 1988; 24 (6): 530-536

    Abstract

    The ability of human epithelial cells derived from adult prostatic tissues to undergo clonal growth in culture was examined. In a previously described serum-free culture system, such cells exhibited a density-dependent growth requirement. It was found that raising the level of one of the constituents of the culture medium, bovine pituitary extract, to 100 micrograms/ml permitted excellent clonal growth when as few as 100 cells were inoculated/60-mm2 dish. Raising the levels of supplements other than pituitary extract (cholera toxin, epidermal growth factor, hydrocortisone, or insulin) did not produce this result. The average colony-forming efficiency of cells derived from primary or early passage cultures was approximately 25%. When single cell suspensions were prepared from tissue isolates and directly analyzed for clonal growth, colony-forming efficiencies were approximately 5%, perhaps indicating the proportion of stem cells with proliferative potential in the original isolates. The colony-forming efficiency of a cell population derived from cancer tissue was not significantly different from those of populations derived from normal tissues.

    View details for Web of Science ID A1988P034100007

    View details for PubMedID 3391931

  • PROSTATE-CANCER - ATTEMPTS TO IDENTIFY DISTINGUISHING PROPERTIES OF MALIGNANT-CELLS FROM INVITRO STUDIES Peehl, D. M., Stamey, T. A. AMER ASSOC CANCER RESEARCH. 1988: 253–253
  • MOLECULAR-BIOLOGY OF PROTOONCOGENES IN GENITOURINARY CANCERS CANCER Peehl, D. M. 1987; 60 (3): 645-649

    Abstract

    The application of molecular biology to oncology has allowed the recognition of altered genes in cancer cells. The DNA sequences most commonly altered belong to the family of proto-oncogenes, which are homologous to the cancer-causing genes of RNA tumor viruses. In the normal cell, proto-oncogenes apparently have important functions in regulation of growth and differentiation. When altered by mutation, deletion, translocation or amplification in cancer cells, proto-oncogenes may disrupt fundamental cellular processes. Such aberrant functioning by abnormal proto-oncogenes may play crucial and even causative roles in cancer development. Altered proto-oncogenes have been identified in cancers of the human urogenital tract. Studies on the expression of proto-oncogenes in genitourinary cells will increase understanding of basic biological properties of these cells, and may yield information relevant to staging, diagnosis, risk factors, and markers of pathologic classification.

    View details for Web of Science ID A1987J281600033

    View details for PubMedID 3594397

  • Oncogene expression. Progress in clinical and biological research Peehl, D. M. 1987; 239: 77-90

    View details for PubMedID 3309967

  • ACTIVATED KI-RAS ONCOGENE IN HUMAN PROSTATIC ADENOCARCINOMA PROSTATE Peehl, D. M., Wehner, N., Stamey, T. A. 1987; 10 (4): 281-289

    Abstract

    The role of cellular oncogenes in the development of human prostate cancer has not been extensively studied. A search for activated oncogenes was undertaken by testing DNA isolated from prostatic adenocarcinoma tissues for transforming activity in a 3T3 transfection assay. A transforming sequence homologous to Ki-ras was detected in one of the samples. DNA from the other cancers was negative in the transformation assay, suggesting that the activation of oncogenes, at least those detectable by the 3T3 transfection assay, is not a frequent event in prostate cancer. Amplification of genomic oncogene sequences in prostatic tissues was also examined, but amplification of Ki-ras, Ha-ras, c-myc, N-myc, c-sis, or c-fos was not detectable in any of the samples.

    View details for Web of Science ID A1987H900700001

    View details for PubMedID 3601807

  • ONCOGENES - A REVIEW WITH RELEVANCE TO CANCERS OF THE UROGENITAL TRACT JOURNAL OF UROLOGY Peehl, D. M., Stamey, T. A. 1986; 135 (5): 897-904

    View details for Web of Science ID A1986C176400001

    View details for PubMedID 3007783

  • SERUM-FREE GROWTH OF ADULT HUMAN PROSTATIC EPITHELIAL-CELLS IN VITRO CELLULAR & DEVELOPMENTAL BIOLOGY Peehl, D. M., Stamey, T. A. 1986; 22 (2): 82-90

    Abstract

    Proliferation of adult human prostatic epithelial cells in serum-free medium occurs upon the addition of cholera toxin, epidermal growth factor, pituitary extract, and hydrocortisone to basal medium PFMR-4A. Insulin and selenium enhance proliferation and permit growth at lower cell densities. Reducing the level of calcium in the medium dramatically alters morphology and also seems to increase proliferation. Mortal strains of cells derived from normal central or peripheral zone, benign hyperplasia, or cancer respond similarly to growth factors and calcium, but two populations of cancer cells which have been long-lived and may be immortal lines behave differently. GKC-CA cells require serum proteins or high levels of pituitary extract for optimal growth, and neither GKC-CA cells or cells of another cancer line, WB-CA, proliferate well in medium containing reduced levels of calcium. These observations may, however, be a reflection of attachment phenomena rather than of growth responses per se. Growth of cells in serum-free medium has allowed definitive studies of the effects of androgens, and regardless of cell type no response to androgens of prostate epithelial cells under any experimental conditions has been seen.

    View details for Web of Science ID A1986A180200004

    View details for PubMedID 2419302

  • GROWTH-RESPONSES OF NORMAL, BENIGN HYPERPLASTIC, AND MALIGNANT HUMAN PROSTATIC EPITHELIAL-CELLS INVITRO TO CHOLERA-TOXIN, PITUITARY EXTRACT, AND HYDROCORTISONE PROSTATE Peehl, D. M., Stamey, T. A. 1986; 8 (1): 51-61

    Abstract

    Normal, benign hyperplastic (BPH), and malignant prostatic epithelial cells from adult humans can be serially passaged in medium PFMR-4 supplemented with 20% whole serum, cholera toxin, and epidermal growth factor (EGF). Improved growth of these cells has been achieved by optimizing the concentrations of the components of the basal medium. In the new modified medium, PFMR-4A, the only supplements required are 5% dialyzed serum, cholera toxin, EGF, pituitary extract, and hydrocortisone. Comparative studies of the growth responses of prostatic cells derived from normal central zone, normal peripheral zone, BPH nodules, and adenocarcinomas did not reveal any qualitative differences; all respond positively to the addition of cholera toxin, pituitary extract, and hydrocortisone to the culture medium. Cells of one cancer strain, however, were much less density-dependent for growth than were the other strains of normal, BPH, or malignant cells.

    View details for Web of Science ID A1986AZB3700005

    View details for PubMedID 2418431

  • KERATIN PROTEIN IN HUMAN PROSTATIC TISSUE AND CELL-CULTURE ANNALS OF THE NEW YORK ACADEMY OF SCIENCES Brawer, M. K., Bostwick, D. G., Peehl, D. M., Stamey, T. A. 1985; 455: 729-731
  • KERATIN IMMUNOREACTIVITY IN THE BENIGN AND NEOPLASTIC HUMAN-PROSTATE CANCER RESEARCH Brawer, M. K., Peehl, D. M., Stamey, T. A., Bostwick, D. G. 1985; 45 (8): 3663-3667

    Abstract

    Keratin immunoreactivity in the benign and neoplastic human prostate was examined immunohistochemically using two monoclonal antibodies with differing specificities. One of these antibodies stained only the basal cells of the normal and hyperplastic prostatic epithelium, with no reactivity in tumor cells of prostatic adenocarcinoma. The other monoclonal antibody recognized a keratin protein present in all normal and hyperplastic columnar (secretory) epithelial cells, as well as in all cancer cells regardless of degree of tumor differentiation. In addition, the second antibody stained acinar and ductal epithelial cells exhibiting premalignant changes. Our findings indicate that keratin immunoreactivity differs among the epithelial cell populations of the human prostate, probably reflecting expression of different keratin proteins. The distinctive patterns of staining obtained with these two antibodies may assist in distinguishing hyperplastic from neoplastic prostatic epithelium, as well as in the recognition of basal cell hyperplasia, transitional cell metaplasia, and premalignant changes.

    View details for Web of Science ID A1985AMU5400041

    View details for PubMedID 2410099

  • Serial propagation of adult human prostatic epithelial cells with cholera toxin. In vitro Peehl, D. M., Stamey, T. A. 1984; 20 (12): 981-986

    Abstract

    Reproducible subculture of adult human prostatic epithelial cells from normal, benign hyperplastic and malignant tissue has been achieved. Cholera toxin is the key component in the culture system, but use of an optimal basal medium (PFMR-4) supplemented with a high level of serum in collagen-coated dishes also improves growth and serial propagation.

    View details for PubMedID 6085065