1
|
Revisiting Histone Deacetylases in Human Tumorigenesis: The Paradigm of Urothelial Bladder Cancer. Int J Mol Sci 2019; 20:ijms20061291. [PMID: 30875794 PMCID: PMC6471041 DOI: 10.3390/ijms20061291] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/24/2022] Open
Abstract
Urinary bladder cancer is a common malignancy, being characterized by substantial patient mortality and management cost. Its high somatic-mutation frequency and molecular heterogeneity usually renders tumors refractory to the applied regimens. Hitherto, methotrexate-vinblastine-adriamycin-cisplatin and gemcitabine-cisplatin represent the backbone of systemic chemotherapy. However, despite the initial chemosensitivity, the majority of treated patients will eventually develop chemoresistance, which severely reduces their survival expectancy. Since chromatin regulation genes are more frequently mutated in muscle-invasive bladder cancer, as compared to other epithelial tumors, targeted therapies against chromatin aberrations in chemoresistant clones may prove beneficial for the disease. “Acetyl-chromatin” homeostasis is regulated by the opposing functions of histone acetyltransferases (HATs) and histone deacetylases (HDACs). The HDAC/SIRT (super-)family contains 18 members, which are divided in five classes, with each family member being differentially expressed in normal urinary bladder tissues. Since a strong association between irregular HDAC expression/activity and tumorigenesis has been previously demonstrated, we herein attempt to review the accumulated published evidences that implicate HDACs/SIRTs as critical regulators in urothelial bladder cancer. Moreover, the most extensively investigated HDAC inhibitors (HDACis) are also analyzed, and the respective clinical trials are also described. Interestingly, it seems that HDACis should be preferably used in drug-combination therapeutic schemes, including radiation.
Collapse
|
2
|
Tanji N, Ozawa A, Kikugawa T, Miura N, Sasaki T, Azuma K, Yokoyama M. Potential of histone deacetylase inhibitors for bladder cancer treatment. Expert Rev Anticancer Ther 2014; 11:959-65. [DOI: 10.1586/era.10.230] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
3
|
Kang HW, Yoon HY, Ha YS, Kim WT, Kim YJ, Yun SJ, Lee SC, Kim WJ. FAM70B as a Novel Prognostic Marker for Cancer Progression and Cancer-Specific Death in Muscle-Invasive Bladder Cancer. Korean J Urol 2012; 53:598-606. [PMID: 23060996 PMCID: PMC3460001 DOI: 10.4111/kju.2012.53.9.598] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 07/06/2012] [Indexed: 11/29/2022] Open
Abstract
Purpose To validate whether FAM70B, which was found in our micro-array profiling as a prognostic marker for cancer survival, could accurately predict prognosis in patients with muscle-invasive bladder cancer (MIBC). Materials and Methods A total of 124 patients with MIBC were enrolled in this study. The FAM70B expression level was analyzed by real-time polymerase chain reaction by using RNA from tumor tissues. The prognostic effect of FAM70B was evaluated by Kaplan-Meier analysis and a multivariate Cox regression model. Results Kaplan-Meier estimates showed a significant difference in progression-free survival (log-rank test, p=0.011) and cancer-specific survival (log-rank test, p=0.017) according to FAM70B gene expression level. By multivariate Cox regression analysis, high FAM70B expression was predictive of cancer progression (hazard ratio [HR], 2.115, p=0.013) and cancer-specific death (HR, 1.925; p=0.033). In the subgroup analysis, high expression of FAM70B was associated with poor cancer-specific survival, progression-free survival, and overall survival in the patients who underwent cystectomy (log-rank test, p=0.013, p=0.036, p=0.005, respectively). In the chemotherapy group, FAM70B expression was associated with cancer-specific survival and progression-free survival (log-rank test, p=0.013, p=0.042, respectively). Moreover, high FAM70B expression was associated with shorter cancer-specific survival in localized or locally advanced tumor stages (log-rank test, p=0.016). Conclusions We confirmed the significance of FAM70B as a prognostic marker in a validation cohort. Therefore, we propose that the FAM70B gene could be used to more precisely predict cancer progression and cancer-specific death in patients with MIBC.
Collapse
Affiliation(s)
- Ho-Won Kang
- Department of Urology, Chungbuk National University College of Medicine, Cheongju, Korea
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Epigenetic biomarkers in prostate cancer: Current and future uses. Cancer Lett 2012; 342:248-56. [PMID: 22391123 DOI: 10.1016/j.canlet.2012.02.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 02/10/2012] [Accepted: 02/11/2012] [Indexed: 12/18/2022]
Abstract
Epigenome alterations are characteristic of nearly all human malignancies and include changes in DNA methylation, histone modifications and microRNAs (miRNAs). However, what induces these epigenetic alterations in cancer is largely unknown and their mechanistic role in prostate tumorigenesis is just beginning to be evaluated. Identification of the epigenetic modifications involved in the development and progression of prostate cancer will not only identify novel therapeutic targets but also prognostic and diagnostic markers. This review will focus on the use of epigenetic modifications as biomarkers for prostate cancer.
Collapse
|
5
|
Kilinc D, Ozdemir O, Ozdemir S, Korgali E, Koksal B, Uslu A, Gultekin YE. Alterations in promoter methylation status of tumor suppressor HIC1, SFRP2, and DAPK1 genes in prostate carcinomas. DNA Cell Biol 2011; 31:826-32. [PMID: 22136354 DOI: 10.1089/dna.2011.1431] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hypermethylated genomic DNA is a common feature in tumoral tissues, although the prevalence of this modification remains poorly understood. We aimed to determine the frequency of five tumor suppressor (TS) genes in prostate cancer and the correlation between promoter hypermethylation of these genes and low and high grade of prostate carcinomas. A total of 30 prostate tumor specimens were investigated for promoter methylation status of TS hypermethylated in cancer 1 (HIC1), death-associated protein kinase 1 (DAPK1), secreted frizzled-related protein 2 (SFRP2), cyclin-dependent kinase inhibitor 2A (p16), and O-6-methylguanine-DNA methyltransferase (MGMT) genes by using bisulfite modifying method. A high frequency of promoter hypermethylation was found in HIC1 (70.9%), SFRP2 (58.3%), and DAPK1 (33.3%) genes in tumor samples that were examined. The current data show high frequency of hypermethylation changes in HIC1, SFRP2, and DAPK1 genes in prostate carcinomas of high Gleason Score (GS).
Collapse
Affiliation(s)
- Devran Kilinc
- Department of Urology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | | | | | | | | | | | | |
Collapse
|
6
|
Marcinkiewicz K, Scotland KB, Boorjian SA, Nilsson EM, Persson JL, Abrahamsson PA, Allegrucci C, Hughes IA, Gudas LJ, Mongan NP. The androgen receptor and stem cell pathways in prostate and bladder cancers (review). Int J Oncol 2011; 40:5-12. [PMID: 21956088 DOI: 10.3892/ijo.2011.1212] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/12/2011] [Indexed: 12/30/2022] Open
Abstract
Bladder cancer is three times more common in men than in women. However, the physiological basis of the male predominance of bladder cancer remains poorly understood. A higher than expected association of prostate and bladder cancers has also been reported which may indicate a common mechanism of carcinogenesis. Consistent with this, androgens and the androgen receptor (AR) play essential roles in prostate carcinogenesis and are believed to play a role in bladder carcinogenesis. There is also evidence implicating cancer stem cells in prostate and bladder cancers. Indeed putative prostate and bladder cancer stem cells share some common molecular features. We highlight key proteins (CD49f, CD133, PTEN, CD44) which are implicated in both prostate and bladder cancers and are enriched in putative prostate and bladder cancer stem cells. We examine published chromatin immuno-precipitation studies analyzing the genome-wide distribution of the AR to identify AR association with, and by inference potential AR-regulation of, these loci. We discuss recent evidence indicating a role for the AR in the splicing of the key urological stem cell protein CD44. We propose a model whereby aberrant AR regulation of these putative stem cell proteins contributes to malignant transformation of prostate and bladder cells. For these reasons we propose that the relationship between androgens and cancer stem cell associated proteins warrants further investigation.
Collapse
|
7
|
Lewandowska J, Bartoszek A. DNA methylation in cancer development, diagnosis and therapy--multiple opportunities for genotoxic agents to act as methylome disruptors or remediators. Mutagenesis 2011; 26:475-87. [PMID: 21551264 DOI: 10.1093/mutage/ger019] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The role of DNA methylation and recently discovered hydroxymethylation in the function of the human epigenome is currently one of the hottest topics in the life sciences. Progress in this field of research has been further accelerated by the discovery that alterations in the methylome are not only associated with key functions of cells and organisms, such as development, differentiation and gene expression, but may underlie a number of human diseases, including cancer. This review describes both well established and more recent observations concerning alterations in the methylome, i.e. the global and local distribution of 5-methylcytosines, involved in its normal functions. Then, the changes in DNA methylation pattern seen in cancer cells are discussed in the context of their utilisation in cancer diagnostics and treatment. On this basis, comparisons are made between natural covalent DNA modification and that induced by genotoxic agents, chemical carcinogens and antitumour drugs as regards their impact on epigenetic mechanisms. The available data suggest that DNA damage by genotoxins can mimic epigenetic markers and in consequence disrupt the proper function of the epigenome. On the other hand, the same processes in cancer cells, e.g. DNA demethylation as a result of DNA methyltransferase blocking or the induction of DNA repair by DNA adducts, may restore the activity of hypermethylated anticancer genes. The observed multiple mechanisms by which genotoxic agents directly affect methylome function suggest that chemical carcinogens act primarily as epigenome disruptors, whereas mutations are secondary events that occur at later stages of cancer development when genome-protecting mechanisms have already been deregulated.
Collapse
Affiliation(s)
- Joanna Lewandowska
- Chemical Faculty, Gdansk University of Technology, ul. Narutowicza 11/12, 80-233 Gdańsk, Poland
| | | |
Collapse
|
8
|
Multiplexed methylation profiles of tumor suppressor genes in bladder cancer. J Mol Diagn 2010; 13:29-40. [PMID: 21227392 DOI: 10.1016/j.jmoldx.2010.11.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 07/19/2010] [Accepted: 07/20/2010] [Indexed: 12/18/2022] Open
Abstract
Changes in DNA methylation of tumor suppressors can occur early in carcinogenesis and are potentially important early indicators of cancer. The objective of this study was to assess the methylation of 25 tumor suppressor genes in bladder cancer using a methylation-specific (MS) multiplex ligation-dependent probe amplification assay (MLPA). Initial analyses in bladder cancer cell lines (n = 14) and fresh-frozen primary bladder tumor specimens (n = 31) supported the panel of genes selected being altered in bladder cancer. The process of MS-MLPA was optimized for its application in body fluids using two independent training and validation sets of urinary specimens (n = 146), including patients with bladder cancer (n = 96) and controls (n = 50). BRCA1 (71.0%), WT1 (38.7%), and RARB (38.7%) were the most frequently methylated genes in bladder tumors, with WT1 methylation being significantly associated with tumor stage (P = 0.011). WT1 and PAX5A were identified as methylated tumor suppressors. In addition, BRCA1, WT1, and RARB were the most frequently methylated genes in urinary specimens. Receiver operating characteristic curve analyses revealed significant diagnostic accuracies in both urinary sets for BRCA1, RARB, and WT1. The novelty of this report relates to applying MS-MLPA, a multiplexed methylation technique, for tumor suppressors in bladder cancer and body fluids. Methylation profiles of tumor suppressor genes were clinically relevant for histopathological stratification of bladder tumors and offered a noninvasive diagnostic strategy for the clinical management of patients affected with uroepithelial neoplasias.
Collapse
|
9
|
Zhang X, Wu M, Xiao H, Lee MT, Levin L, Leung YK, Ho SM. Methylation of a single intronic CpG mediates expression silencing of the PMP24 gene in prostate cancer. Prostate 2010; 70:765-76. [PMID: 20054818 PMCID: PMC2857536 DOI: 10.1002/pros.21109] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND We previously demonstrated that a putative anti-tumor gene, peroxisomal membrane protein 4, 24 kDa (PMP24 or PXMP4), is silenced via DNA methylation of a CpG island in its 5' flanking region (5'-CGI) in prostate cancer (PCa) cells. METHODS To identify demethylation hypersensitive site(s) in PMP24 5'-CGI, PC-3 cells with methylated 5'-CGI were treated with a low-dose of 5-aza-2'-deoxycytidine (5-aza-dC) just sufficient to reactivate gene expression, referred as the limited demethylation approach. Gel shift assays and promoter analyzes were performed to demonstrate the role of the hypersensitive site in PMP24 gene regulation. Transfection of a methylated oligonucleotide corresponding to the hypersensitive site was conducted to determine the effect of site-specific methylation on the gene expression. Bisulfite sequencing analysis was performed to reveal the methylation status of PMP24 promoter in cultured cells and microdissected samples. In situ hybridization was applied to determine expression positivity of PMP24 mRNA. RESULTS A 5-aza-dC hypersensitive site encompasses two CpG dinucleotides in intron 1 was identified. Methylation of the first, but not the second, CpG dinucleotide of this site disrupted DNA-protein interactions and suppressed the gene expression. Using archival specimens, we found the first CpG dinucleotide of the hypersensitive site is hypermethylated with a loss of PMP24 mRNA expression in microdissected PCa cells when compared to normal prostatic epithelial cells. CONCLUSIONS These findings support a critical role for a single intronic CpG dinucleotide in PMP24 gene regulation through DNA methylation. The data suggest that methylation-mediated silencing of PMP24 is a molecular event associated with prostate carcinogenesis.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Cincinnati Cancer Consortium, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Hong Xiao
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ming-Tsung Lee
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Linda Levin
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yuet-Kin Leung
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Cincinnati Cancer Consortium, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Shuk-Mei Ho
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Cincinnati Cancer Consortium, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Correspondence to: Shuk-Mei Ho, Department of Environmental Health, University of Cincinnati, College of Medicine, Kettering Laboratory, Suite 128, 3223 Eden Ave., Cincinnati, OH 45267. Telephone: (513)-558-5701. Fax: (513)-558-0071.
| |
Collapse
|
10
|
Liu X, Gomez-Pinillos A, Liu X, Johnson EM, Ferrari AC. Induction of bicalutamide sensitivity in prostate cancer cells by an epigenetic Puralpha-mediated decrease in androgen receptor levels. Prostate 2010; 70:179-89. [PMID: 19790234 DOI: 10.1002/pros.21051] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Increased androgen receptor (AR) levels support resistance to apoptosis and hormone therapy in advanced prostate cancer (PC). We recently linked the overexpression of AR in androgen-independent LNCaP cells (AI-cells) and tissues from castration-resistant patients to decreased nuclear levels of Pur-alpha (Puralpha) and loss from a protein complex bound to repressor sequences (ARS) in the 5'-UTR of AR. Strategies to regain control of increased AR transcription may overcome resistance of AI-cells and improve treatment outcomes. METHODS MTT, real-time PCR, Western blot, ChIP, flow cytometry, and caspase 3/7 activation measured the effect on growth and targets of LBH589/bicalutamide treatment of AI-cells and androgen-dependent LNCaP cells (AD). RESULTS Within 16 hr of treatment of AI-cells with low concentrations of the histone deacetylase inhibitor LBH589, a shift of cytoplasmic Puralpha restored the nuclear levels and the binding of Puralpha to the ARS. This was followed by a decline in AR-mRNA and protein reaching levels of parental AD-cells. The fraction of AI-cells in G1 increased and the cells in S phase decreased similar to AD-cells, and there was a modest caspase activation. Most notably, treatment of bicalutamide-resistant AI-cells with 10 nM LBH589 combined with 12.5 microM bicalutamide synergistically inhibited cell growth and induced a fivefold higher level of caspase 3/7 activation than observed in AD-cells. CONCLUSIONS Low-dose LBH589 restores Puralpha binding to ARS and down-regulates AR transcription. Biologically, LBH589 reverses the resistance of AI-cells to bicalutamide and to apoptosis. The combination may restore the hormonal response of castration-resistant PC patients.
Collapse
Affiliation(s)
- Xiaomei Liu
- New York University Cancer Institute and Department of Medicine, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
11
|
Aleman A, Cebrian V, Alvarez M, Lopez V, Orenes E, Lopez-Serra L, Algaba F, Bellmunt J, López-Beltrán A, Gonzalez-Peramato P, Cordon-Cardo C, García J, del Muro JG, Esteller M, Sánchez-Carbayo M. Identification of PMF1 methylation in association with bladder cancer progression. Clin Cancer Res 2009; 14:8236-43. [PMID: 19088041 DOI: 10.1158/1078-0432.ccr-08-0778] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Polyamines are important regulators of cell growth and death. The polyamine modulated factor-1 (PMF-1) is involved in polyamine homeostasis. After identifying an enriched CpG island encompassing the PMF1 promoter, we aimed at evaluating the clinical relevance of PMF1 methylation in bladder cancer. EXPERIMENTAL DESIGN The epigenetic silencing of PMF1 by hypermethylation was tested in bladder cancer cells (n = 11) after azacytidine treatment. PMF1 methylation status was evaluated in 507 bladder tumors and 118 urinary specimens of bladder cancer patients and controls. PMF1 protein expression was analyzed by immunohistochemistry on tissue arrays containing bladder tumors for which PMF1 methylation was assessed (n = 218). RESULTS PMF1 hypermethylation was associated with gene expression loss, being restored in vitro by a demethylating agent. An initial set of 101 primary frozen bladder tumors served to identify PMF1 hypermethylation in 88.1% of the cases. An independent set of 406 paraffin-embedded tumors also revealed a high PMF1 methylation rate (77.6%). PMF1 methylation was significantly associated with increasing stage (P = 0.025). Immunohistochemical analyses revealed that PMF1 methylation was associated with cytoplasmic PMF1 expression loss (P = 0.032). PMF1 protein expression patterns were significantly associated with stage (P < 0.001), grade (P < 0.001), and poor overall survival using univariate (P < 0.001) and multivariate (P = 0.011) analyses. Moreover, PMF1 methylation in urinary specimens distinguished bladder cancer patients from controls (area under the curve = 0.800). CONCLUSION PMF1 was identified to be epigenetically modified in bladder cancer. The association of PMF1 methylation with tumor progression and its diagnostic ability using urinary specimens support including PMF1 assessment for the clinical management of bladder cancer patients.
Collapse
Affiliation(s)
- Ainel Aleman
- Tumor Markers Group, Molecular Pathology Program, Spanish National Cancer Center, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Chiam K, Tilley WD, Butler LM, Bianco-Miotto T. The dynamic and static modification of the epigenome by hormones: a role in the developmental origin of hormone related cancers. Biochim Biophys Acta Rev Cancer 2009; 1795:104-9. [PMID: 19166905 DOI: 10.1016/j.bbcan.2008.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 12/14/2008] [Accepted: 12/21/2008] [Indexed: 12/19/2022]
Abstract
There are numerous diseases associated with abnormal hormonal regulation and these include cancers of the breast and prostate. There is substantial evidence that early hormonal perturbations (in utero or during early development) are associated with increased disease susceptibility later in life. These perturbations may arise from exposure to environmental agents or endocrine disruptors which mimic hormones and disrupt normal hormonal signaling. Epigenetic alterations have often been proposed as the underlying mechanism by which early hormonal perturbations may give rise to disease in adulthood. Currently, there is minimal evidence to support a direct link between early hormonal perturbations and epigenetic modifications; or between epigenetic alterations and subsequent onset of cancer. Given that epigenetic modifications may play an important role in hormone-dependent cancers, it is essential to better understand the relationship between the hormonal environment and epigenetic modifications in both normal and disease states. In this review, we highlight several important studies which support the hypothesis that: hormonal perturbations early in life may result in epigenetic changes that may modify hormone receptor function, thereby contributing to an increased risk of developing hormone-related cancers.
Collapse
Affiliation(s)
- Karen Chiam
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide, Hanson Institute, PO Box 14 Rundle Mall, Adelaide, South Australia, 5000, Australia
| | | | | | | |
Collapse
|
13
|
Di Maro S, Pong RC, Hsieh JT, Ahn JM. Efficient Solid-Phase Synthesis of FK228 Analogues as Potent Antitumoral Agents. J Med Chem 2008; 51:6639-41. [DOI: 10.1021/jm800959f] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Salvatore Di Maro
- Department of Chemistry, University of Texas at Dallas, Richardson, Texas 75080, and Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Rey-Chen Pong
- Department of Chemistry, University of Texas at Dallas, Richardson, Texas 75080, and Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Jer-Tsong Hsieh
- Department of Chemistry, University of Texas at Dallas, Richardson, Texas 75080, and Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Jung-Mo Ahn
- Department of Chemistry, University of Texas at Dallas, Richardson, Texas 75080, and Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| |
Collapse
|
14
|
|
15
|
Hassig CA, Symons KT, Guo X, Nguyen PM, Annable T, Wash PL, Payne JE, Jenkins DA, Bonnefous C, Trotter C, Wang Y, Anzola JV, Milkova EL, Hoffman TZ, Dozier SJ, Wiley BM, Saven A, Malecha JW, Davis RL, Muhammad J, Shiau AK, Noble SA, Rao TS, Smith ND, Hager JH. KD5170, a novel mercaptoketone-based histone deacetylase inhibitor that exhibits broad spectrum antitumor activity in vitro and in vivo. Mol Cancer Ther 2008; 7:1054-65. [PMID: 18483295 DOI: 10.1158/1535-7163.mct-07-2347] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Histone deacetylase (HDAC) inhibitors have garnered significant attention as cancer drugs. These therapeutic agents have recently been clinically validated with the market approval of vorinostat (SAHA, Zolinza) for treatment of cutaneous T-cell lymphoma. Like vorinostat, most of the small-molecule HDAC inhibitors in clinical development are hydroxamic acids, whose inhibitory activity stems from their ability to coordinate the catalytic Zn2+ in the active site of HDACs. We sought to identify novel, nonhydroxamate-based HDAC inhibitors with potentially distinct pharmaceutical properties via an ultra-high throughput small molecule biochemical screen against the HDAC activity in a HeLa cell nuclear extract. An alpha-mercaptoketone series was identified and chemically optimized. The lead compound, KD5170, exhibits HDAC inhibitory activity with an IC50 of 0.045 micromol/L in the screening biochemical assay and an EC50 of 0.025 micromol/L in HeLa cell-based assays that monitor histone H3 acetylation. KD5170 also exhibits broad spectrum classes I and II HDAC inhibition in assays using purified recombinant human isoforms. KD5170 shows significant antiproliferative activity against a variety of human tumor cell lines, including the NCI-60 panel. Significant tumor growth inhibition was observed after p.o. dosing in human HCT-116 (colorectal cancer), NCI-H460 (non-small cell lung carcinoma), and PC-3 (prostate cancer) s.c. xenografts in nude mice. In addition, a significant increase in antitumor activity and time to end-point occurred when KD5170 was combined with docetaxel in xenografts of the PC-3 prostate cancer cell line. The biological and pharmaceutical profile of KD5170 supports its continued preclinical and clinical development as a broad spectrum anticancer agent.
Collapse
|
16
|
Weichert W, Röske A, Gekeler V, Beckers T, Stephan C, Jung K, Fritzsche FR, Niesporek S, Denkert C, Dietel M, Kristiansen G. Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer 2008; 98:604-10. [PMID: 18212746 PMCID: PMC2243142 DOI: 10.1038/sj.bjc.6604199] [Citation(s) in RCA: 366] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High activity of histone deacetylases (HDACs) causes epigenetic alterations associated with malignant cell behaviour. Consequently, HDAC inhibitors have entered late-phase clinical trials as new antineoplastic drugs. However, little is known about expression and function of specific HDAC isoforms in human tumours including prostate cancer. We investigated the expression of class I HDACs in 192 prostate carcinomas by immunohistochemistry and correlated our findings to clinicopathological parameters including follow-up data. Class I HDAC isoforms were strongly expressed in the majority of the cases (HDAC1: 69.8%, HDAC2: 74%, HDAC3: 94.8%). High rates of HDAC1 and HDAC2 expression were significantly associated with tumour dedifferentiation. Strong expression of all HDACs was accompanied by enhanced tumour cell proliferation. In addition, HDAC2 was an independent prognostic marker in our prostate cancer cohort. In conclusion, we showed that the known effects of HDACs on differentiation and proliferation of cancer cells observed in vitro can also be confirmed in vivo. The class I HDAC isoforms 1, 2 and 3 are differentially expressed in prostate cancer, which might be important for upcoming studies on HDAC inhibitors in this tumour entity. Also, the highly significant prognostic value of HDAC2 clearly deserves further study.
Collapse
Affiliation(s)
- W Weichert
- Institute of Pathology, Charité - Universitätsmedizin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Valdespino V, Tsagozis P, Pisa P. Current perspectives in the treatment of advanced prostate cancer. Med Oncol 2008; 24:273-86. [PMID: 17873302 DOI: 10.1007/s12032-007-0017-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 11/30/1999] [Accepted: 01/09/2007] [Indexed: 01/26/2023]
Abstract
Prostate cancer (PC) continues to be an important world health problem for men. Patients with locally confined PC are treated with either radiotherapy or surgery. However, treatment of more advanced stages of the disease is problematic. Initially, androgen deprivation offers a period of clinical stability, which is however invariably followed by progression to non-responsiveness to hormonal manipulation. Current management of patients with androgen-independent prostate cancer (AIPC) displays modest response rates and achieves only short-term benefit. Recently, knowledge in the complex pathophysiology of advanced PC has led to the identification of mechanisms and target molecules permitting the introduction of new therapies. Consequently, many investigational treatments are ongoing for AIPC in Phase-II and Phase-III trials aiming at the combination of chemotherapeutic regimens along with immunotherapy targeting PC-associated antigens. Other attractive options are gene therapy, as well as the targeting of survival signaling, differentiation, and apoptosis of the malignant PC cells. Further treatment modalities are directed against the tumor microenvironment, bone metastasis, or both. Collectively, the aforementioned efforts introduce a new era in the management of advanced PC. Novel pharmaceutical compounds and innovative approaches, integrated into the concept of individualized therapy will hopefully, during the next decade, improve the outcome and survival for hundreds of thousands of men worldwide.
Collapse
Affiliation(s)
- Victor Valdespino
- Department of Surgery, UMAE de Oncologia del CMN SXXI, Instituto Mexicano del Seguro Social, Universidad Autonoma Metropolitana, Mexico, Mexico
| | | | | |
Collapse
|
18
|
Zhang Z, Stanfield J, Frenkel E, Kabbani W, Hsieh JT. Enhanced therapeutic effect on androgen-independent prostate cancer by depsipeptide (FK228), a histone deacetylase inhibitor, in combination with docetaxel. Urology 2007; 70:396-401. [PMID: 17826527 DOI: 10.1016/j.urology.2007.03.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Revised: 02/28/2007] [Accepted: 03/16/2007] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To study whether a histone deacetylase inhibitor, FK228, can enhance therapeutic efficacies of other chemotherapeutic agents against androgen-independent prostate cancer (AIPCa) and to explore any possible early biomarker for predicting drug response. METHODS The therapeutic effects of FK228-based combinations on three AIPCa cell lines (PC-3, DU145, and C4-2) were evaluated by synergisms of cytotoxicity. Cell cycle analysis and Western blot assay were used to study the underlying mechanisms of drug action and search for any potential surrogate biomarker. In addition, we investigated the in vivo antitumor effects of the FK228/docetaxel combination using the PC-3 xenograft model. RESULTS FK228/docetaxel surpassed other FK228-based combinations by achieving more synergism of cytotoxicity. FK228 enhanced the therapeutic effect of docetaxel against AIPCa by exhibiting markedly enhanced and prolonged inhibitory effects in vitro and better tumor regression in vivo by inducing apoptosis. It appears that p21(WAF1) induction consistently correlates with single or combination treatment. CONCLUSIONS The results of our study have shown that FK228 is able to enhance the therapeutic effect of docetaxel against AIPCa. FK228 appears to be a promising second-line option in combination with docetaxel. The p21(WAF1) protein level can be used as a surrogate biomarker to predict and monitor the therapeutic response.
Collapse
Affiliation(s)
- Zhengwang Zhang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9110, USA
| | | | | | | | | |
Collapse
|
19
|
Blank-Porat D, Gruss-Fischer T, Tarasenko N, Malik Z, Nudelman A, Rephaeli A. The anticancer prodrugs of butyric acid AN-7 and AN-9, possess antiangiogenic properties. Cancer Lett 2007; 256:39-48. [PMID: 17611019 DOI: 10.1016/j.canlet.2007.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 05/10/2007] [Accepted: 05/18/2007] [Indexed: 11/28/2022]
Abstract
The antiangiogenic and antineoplastic activities of the butyric acid prodrugs AN-7 and AN-9 were demonstrated in vitro with HUVEC by inhibition of proliferation and vascular tubes formation, enhanced apoptosis, and inhibition of 22Rv-1 cells migration. In the sc implanted human prostate tumors (22Rv-1) in nude mice, AN-7 significantly inhibited Ki-67, HIF-1alpha, HER-2/neu, bFGF and increased PTEN level. AN-7 and AN-9 reduced hemoglobin accumulation in matrigel plugs implanted sc in Balb-c mice. Herein, we show that the anticancer activity of AN-7 and AN-9 can be attributed in part to their antiangiogenic activities suggesting potential therapeutic benefits for prostate cancer patients.
Collapse
Affiliation(s)
- Diana Blank-Porat
- Felsenstein Center for Medical Research, Tel Aviv University Beilinson Campus, Petach Tikva 49100, Israel
| | | | | | | | | | | |
Collapse
|
20
|
Bourdeau-Heller J, Oberley TD. Prostate carcinoma cells selected by long-term exposure to reduced oxygen tension show remarkable biochemical plasticity via modulation of superoxide, HIF-1alpha levels, and energy metabolism. J Cell Physiol 2007; 212:744-52. [PMID: 17458899 DOI: 10.1002/jcp.21069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cancer cells are able to tolerate levels of O(2) that are damaging or lethal to normal cells; we hypothesize that this tolerance is the result of biochemical plasticity which maintains cellular homeostasis of both energy levels and oxidation state. In order to examine this hypothesis, we used different O(2) levels as a selective agent during long-term culture of DU145 prostate cancer cells to develop three isogenic cell lines that grow in normoxic (4%), hyperoxic (21%), or hypoxic (1%) O(2) conditions. Growth characteristics and O(2) consumption differed significantly between these cell lines without changes in ATP levels or altered sensitivity to 2-deoxy-D-glucose, an inhibitor of glycolysis. O(2) consumption was significantly higher in the hyperoxic line as was the level of endogenous superoxide. The hypoxic cell line regulated the chemical gradient of the proton motive force (PMF) independent of the electrical component without O(2)-dependent changes in Hif-1alpha levels. In contrast, the normoxic line regulated Hif-1alpha without tight regulation of the chemical component of the PMF noted in the hypoxic cell line. From these studies, we conclude that selection of prostate cancer cells by long-term exposure to low ambient levels of O(2) resulted in cells with unique biochemical properties in which energy metabolism, reactive oxygen species (ROS), and HIF-1alpha levels are modulated to allow cell survival and growth. Thus, cancer cells exhibit remarkable biochemical plasticity in response to various O(2) levels.
Collapse
Affiliation(s)
- Jeanne Bourdeau-Heller
- University of Wisconsin School of Medicine and Public Health, Department of Pathology, Madison, WI 53705, USA
| | | |
Collapse
|
21
|
Abstract
Ageing of the male reproductive system is characterized by changes in the endocrine system, hypogonadism, erectile dysfunction and proliferative disorders of the prostate gland. Stochastic damage accumulating within ageing leads to progressive dysregulation at each level of the hypothalamic-pituitary-gonadal (HPG) axis and in local auto/paracrine interactions, thereby inducing morphological changes in reproductive target organs, such as the prostate, testis and penis. Despite age-related changes in the HPG axis, endocrine functions are generally sufficient to maintain fertility in elderly men. Ageing of the male reproductive system can give rise to clinically relevant manifestations, such as benign prostatic hyperplasia (BPH), prostate cancer (PCa) and erectile dysfunction (ED). In this review, we discuss morphological/histological changes occurring in these organs and current views and concepts of the underlying pathology. Moreover, we emphasize the molecular/cellular pathways leading to reduced testicular/penile function and proliferative disorders of the prostate gland.
Collapse
Affiliation(s)
- N Sampson
- Institute for Biomedical Ageing Research, Austrian Academy of Sciences, Innsbruck, Austria
| | | | | | | |
Collapse
|