151
|
Tsai H, Morais CL, Alshalalfa M, Tan HL, Haddad Z, Hicks J, Gupta N, Epstein JI, Netto GJ, Isaacs WB, Luo J, Mehra R, Vessella RL, Karnes RJ, Schaeffer EM, Davicioni E, De Marzo AM, Lotan TL. Cyclin D1 Loss Distinguishes Prostatic Small-Cell Carcinoma from Most Prostatic Adenocarcinomas. Clin Cancer Res 2015; 21:5619-29. [PMID: 26246306 DOI: 10.1158/1078-0432.ccr-15-0744] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/26/2015] [Indexed: 02/02/2023]
Abstract
PURPOSE Small-cell neuroendocrine differentiation in prostatic carcinoma is an increasingly common resistance mechanism to potent androgen deprivation therapy (ADT), but can be difficult to identify morphologically. We investigated whether cyclin D1 and p16 expression can inform on Rb functional status and distinguish small-cell carcinoma from adenocarcinoma. EXPERIMENTAL DESIGN We used gene expression data and immunohistochemistry to examine cyclin D1 and p16 levels in patient-derived xenografts (PDX), and prostatic small-cell carcinoma and adenocarcinoma specimens. RESULTS Using PDX, we show proof-of-concept that a high ratio of p16 to cyclin D1 gene expression reflects underlying Rb functional loss and distinguishes morphologically identified small-cell carcinoma from prostatic adenocarcinoma in patient specimens (n = 13 and 9, respectively). At the protein level, cyclin D1, but not p16, was useful to distinguish small-cell carcinoma from adenocarcinoma. Overall, 88% (36/41) of small-cell carcinomas showed cyclin D1 loss by immunostaining compared with 2% (2/94) of Gleason score 7-10 primary adenocarcinomas at radical prostatectomy, 9% (4/44) of Gleason score 9-10 primary adenocarcinomas at needle biopsy, and 7% (8/115) of individual metastases from 39 patients at autopsy. Though rare adenocarcinomas showed cyclin D1 loss, many of these were associated with clinical features of small-cell carcinoma, and in a cohort of men treated with adjuvant ADT who developed metastasis, lower cyclin D1 gene expression was associated with more rapid onset of metastasis and death. CONCLUSIONS Cyclin D1 loss identifies prostate tumors with small-cell differentiation and may identify a small subset of adenocarcinomas with poor prognosis. Clin Cancer Res; 21(24); 5619-29. ©2015 AACR.
Collapse
Affiliation(s)
- Harrison Tsai
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carlos L Morais
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Hsueh-Li Tan
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zaid Haddad
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Jessica Hicks
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nilesh Gupta
- Pathology, Henry Ford Health System, Detroit, Michigan
| | - Jonathan I Epstein
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George J Netto
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William B Isaacs
- Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Luo
- Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rohit Mehra
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan
| | - Robert L Vessella
- Department of Urology, University of Washington, Seattle, Washington
| | | | - Edward M Schaeffer
- Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elai Davicioni
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Angelo M De Marzo
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tamara L Lotan
- Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
152
|
Oser MG, Niederst MJ, Sequist LV, Engelman JA. Transformation from non-small-cell lung cancer to small-cell lung cancer: molecular drivers and cells of origin. Lancet Oncol 2015; 16:e165-72. [PMID: 25846096 DOI: 10.1016/s1470-2045(14)71180-5] [Citation(s) in RCA: 665] [Impact Index Per Article: 73.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung cancer is the most common cause of cancer deaths worldwide. The two broad histological subtypes of lung cancer are small-cell lung cancer (SCLC), which is the cause of 15% of cases, and non-small-cell lung cancer (NSCLC), which accounts for 85% of cases and includes adenocarcinoma, squamous-cell carcinoma, and large-cell carcinoma. Although NSCLC and SCLC are commonly thought to be different diseases owing to their distinct biology and genomic abnormalities, the idea that these malignant disorders might share common cells of origin has been gaining support. This idea has been supported by the unexpected findings that a subset of NSCLCs with mutated EGFR return as SCLC when resistance to EGFR tyrosine kinase inhibitors develops. Additionally, other case reports have described the coexistence of NSCLC and SCLC, further challenging the commonly accepted view of their distinct lineages. Here, we summarise the published clinical observations and biology underlying tumours with combined SCLC and NSCLC histology and cancers that transform from adenocarcinoma to SCLC. We also discuss pre-clinical studies pointing to common potential cells of origin, and speculate how the distinct paths of differentiation are determined by the genomics of each disease.
Collapse
Affiliation(s)
- Matthew G Oser
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA
| | - Matthew J Niederst
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center, Department of Medicine and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
153
|
Zhang X, Coleman IM, Brown LG, True LD, Kollath L, Lucas JM, Lam HM, Dumpit R, Corey E, Chéry L, Lakely B, Higano CS, Montgomery B, Roudier M, Lange PH, Nelson PS, Vessella RL, Morrissey C. SRRM4 Expression and the Loss of REST Activity May Promote the Emergence of the Neuroendocrine Phenotype in Castration-Resistant Prostate Cancer. Clin Cancer Res 2015; 21:4698-708. [PMID: 26071481 DOI: 10.1158/1078-0432.ccr-15-0157] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 05/14/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE The neuroendocrine phenotype is associated with the development of metastatic castration-resistant prostate cancer (CRPC). Our objective was to characterize the molecular features of the neuroendocrine phenotype in CRPC. EXPERIMENTAL DESIGN Expression of chromogranin A (CHGA), synaptophysin (SYP), androgen receptor (AR), and prostate-specific antigen (PSA) was analyzed by IHC in 155 CRPC metastases from 50 patients and in 24 LuCaP prostate cancer patient-derived xenografts (PDX). Seventy-one of 155 metastases and the 24 LuCaP xenograft lines were analyzed by whole-genome microarrays. REST splicing was verified by PCR. RESULTS Coexpression of CHGA and SYP in >30% of cells was observed in 22 of 155 metastases (9 patients); 11 of the 22 metastases were AR(+)/PSA(+) (6 patients), 11/22 were AR-/PSA- (4 patients), and 4/24 LuCaP PDXs were AR(-)/PSA(-). By IHC, of the 71 metastases analyzed by whole-genome microarrays, 5 metastases were CHGA(+)/SYP(+)/AR(-), and 5 were CHGA(+)/SYP(+)/AR(+). Only CHGA(+)/SYP(+) metastases had a neuroendocrine transcript signature. The neuronal transcriptional regulator SRRM4 transcript was associated with the neuroendocrine signature in CHGA(+)/SYP(+) metastases and all CHGA(+)/SYP(+) LuCaP xenografts. In addition, expression of SRRM4 in LuCaP neuroendocrine xenografts correlated with a splice variant of REST that lacks the transcriptional repressor domain. CONCLUSIONS (i) Metastatic neuroendocrine status can be heterogeneous in the same patient, (ii) the CRPC neuroendocrine molecular phenotype can be defined by CHGA(+)/SYP(+) dual positivity, (iii) the neuroendocrine phenotype is not necessarily associated with the loss of AR activity, and (iv) the splicing of REST by SRRM4 could promote the neuroendocrine phenotype in CRPC.
Collapse
Affiliation(s)
- Xiaotun Zhang
- Department of Urology, University of Washington, Seattle, Washington
| | - Ilsa M Coleman
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lisha G Brown
- Department of Urology, University of Washington, Seattle, Washington
| | - Lawrence D True
- Department of Pathology, University of Washington, Seattle, Washington
| | - Lori Kollath
- Department of Urology, University of Washington, Seattle, Washington
| | - Jared M Lucas
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, Washington
| | - Ruth Dumpit
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Lisly Chéry
- Department of Urology, University of Washington, Seattle, Washington
| | - Bryce Lakely
- Department of Urology, University of Washington, Seattle, Washington
| | - Celestia S Higano
- Department of Urology, University of Washington, Seattle, Washington. Department of Medicine, University of Washington, Seattle, Washington
| | - Bruce Montgomery
- Department of Medicine, University of Washington, Seattle, Washington
| | - Martine Roudier
- Department of Urology, University of Washington, Seattle, Washington
| | - Paul H Lange
- Department of Urology, University of Washington, Seattle, Washington. Department of Veterans Affairs Medical Center, Seattle, Washington
| | - Peter S Nelson
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Medicine, University of Washington, Seattle, Washington
| | - Robert L Vessella
- Department of Urology, University of Washington, Seattle, Washington. Department of Veterans Affairs Medical Center, Seattle, Washington
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington.
| |
Collapse
|
154
|
Ferraldeschi R, Welti J, Luo J, Attard G, de Bono JS. Targeting the androgen receptor pathway in castration-resistant prostate cancer: progresses and prospects. Oncogene 2015; 34:1745-57. [PMID: 24837363 PMCID: PMC4333106 DOI: 10.1038/onc.2014.115] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/24/2014] [Accepted: 03/24/2014] [Indexed: 12/11/2022]
Abstract
Androgen receptor (AR) signaling is a critical pathway for prostate cancer cells, and androgen-deprivation therapy (ADT) remains the principal treatment for patients with locally advanced and metastatic disease. However, over time, most tumors become resistant to ADT. The view of castration-resistant prostate cancer (CRPC) has changed dramatically in the last several years. Progress in understanding the disease biology and mechanisms of castration resistance led to significant advancements and to paradigm shift in the treatment. Accumulating evidence showed that prostate cancers develop adaptive mechanisms for maintaining AR signaling to allow for survival and further evolution. The aim of this review is to summarize molecular mechanisms of castration resistance and provide an update in the development of novel agents and strategies to more effectively target the AR signaling pathway.
Collapse
Affiliation(s)
- R Ferraldeschi
- Prostate Cancer Targeted Therapy Group, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - J Welti
- Prostate Cancer Targeted Therapy Group, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - J Luo
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G Attard
- Prostate Cancer Targeted Therapy Group, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - JS de Bono
- Prostate Cancer Targeted Therapy Group, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| |
Collapse
|
155
|
Gordon CA, Gulzar ZG, Brooks JD. NUSAP1 expression is upregulated by loss of RB1 in prostate cancer cells. Prostate 2015; 75:517-26. [PMID: 25585568 DOI: 10.1002/pros.22938] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/05/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Overexpression of NUSAP1 is associated with poor prognosis in prostate cancer, but little is known about what leads to its overexpression. Based on previous observations that NUSAP1 expression is enhanced by E2F1, we hypothesized that NUSAP1 expression is regulated, at least in part, by loss of RB1 via the RB1/E2F1 axis. METHODS Using Significance Analysis of Microarrays, we examined RB1, E2F1, and NUSAP1 transcript levels in prostate cancer gene expression datasets. We compared NUSAP1 expression levels in DU145, LNCaP, and PC-3 prostate cancer cell lines via use of cDNA microarray data, RT-qPCR, and Western blots. In addition, we used lentiviral expression constructs to knockdown RB1 in prostate cancer cell lines and transient transfections to knockdown E2F1, and investigated RB1, E2F1, and NUSAP1 expression levels with RT-qPCR and Western blots. Finally, in DU145 cells or PC-3 cells that stably underexpress RB1, we used proliferation and invasion assays to assess whether NUSAP1 knockdown affects proliferation or invasion. RESULTS NUSAP1 transcript levels are positively correlated with E2F1 and negatively correlated with RB1 transcript levels in prostate cancer microarray datasets. NUSAP1 expression is elevated in the RB1-null DU145 prostate cancer cell line, as opposed to LNCaP and PC-3 cell lines. Furthermore, NUSAP1 expression increases upon knockdown of RB1 in prostate cancer cell lines (LNCaP and PC-3) and decreases after knockdown of E2F1. Lastly, knockdown of NUSAP1 in DU145 cells or PC-3 cells with stable knockdown of RB1 decreases proliferation and invasion of these cells. CONCLUSION Our studies support the notion that NUSAP1 expression is upregulated by loss of RB1 via the RB1/E2F1 axis in prostate cancer cells. Such upregulation may promote prostate cancer progression by increasing proliferation and invasion of prostate cancer cells. NUSAP1 may thus represent a novel therapeutic target.
Collapse
Affiliation(s)
- Catherine A Gordon
- Department of Urology, Stanford University School of Medicine, Stanford, California
| | | | | |
Collapse
|
156
|
Yadav SS, Li J, Lavery HJ, Yadav KK, Tewari AK. Next-generation sequencing technology in prostate cancer diagnosis, prognosis, and personalized treatment. Urol Oncol 2015; 33:267.e1-13. [PMID: 25791755 DOI: 10.1016/j.urolonc.2015.02.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 02/06/2023]
Abstract
Next-generation sequencing (NGS) of the genetic information of cancer cells has revolutionized the field of cancer biology, including prostate cancer (PCa). New recurrent alterations have been identified in PCa (e.g., TMPRSS2-ERG translocation, SPOP and CHD1 mutations, and chromoplexy), and many previous ones in well-established pathways have been validated (e.g., androgen receptor overexpression and mutations; PTEN, RB1, and TP53 loss/mutations). With its highly heterogeneous nature, PCa continues to pose a tremendous challenge in terms of diagnosis and prognosis. Combining the information gained through NGS studies with clinicopathological and radiological data will help diagnose the aggressiveness of the cancer with greater accuracy. Furthermore, understanding the heterogeneity of tumor through single-cell or single-molecule sequencing technology will also strengthen the prognosis and provide better, patient-specific drug identification. As this research becomes more prominent, it is important that urologic oncologists become familiar with the various NGS technologies and the results generated using them. We highlight the commonly used NGS tools and summarize recent discoveries relevant to PCa.
Collapse
Affiliation(s)
- Shalini S Yadav
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY
| | - Jinyi Li
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY
| | - Hugh J Lavery
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY
| | - Kamlesh K Yadav
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY.
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY.
| |
Collapse
|
157
|
Inactivation of the retinoblastoma gene yields a mouse model of malignant colorectal cancer. Oncogene 2015; 34:5890-9. [PMID: 25745996 PMCID: PMC4668801 DOI: 10.1038/onc.2015.30] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 11/22/2014] [Accepted: 11/25/2014] [Indexed: 02/07/2023]
Abstract
The retinoblastoma gene (Rb) is mutated at significant frequency in various human epithelial tumors, including colorectal cancer, and is strongly associated with metastatic disease. However, sole inactivation of Rb in the mouse has so far failed to yield epithelial cancers. Here, we specifically inactivate Rb and/or p53 in the urogenital epithelium and the intestine. We find that loss of both tumor suppressors is unable to yield tumors in the transitional epithelium lining the bladder, kidneys and ureters. Instead, these mice develop highly metastatic tumors of neuroendocrine, not epithelial, origin within the urogenital tract to give prostate cancer in the males and vaginal tumors in the females. Additionally, we discovered that the sole inactivation of Rb in the intestine was sufficient to induce formation of metastatic colorectal adenocarcinomas. These tumors closely mirror the human disease in regard to age of onset, histological appearance, invasiveness and metastatic potential. Like most human colorectal carcinomas, our murine Rb-deficient tumors demonstrate genomic instability and they show activation of β-catenin. Deregulation of the Wnt/β-catenin pathway is specific to the intestinal tumors, as genomic instability but not activation of β-catenin was observed in the neuroendocrine tumors. To date, attempts to generate genetically engineered mouse models of colorectal cancer tumors have yielded mostly cancer of the small intestine, which rarely occurs in humans. Our system provides the opportunity to accurately model and study colorectal cancer in the mouse via a single gene mutation.
Collapse
|
158
|
de Leeuw R, Berman-Booty LD, Schiewer MJ, Ciment SJ, Den RB, Dicker AP, Kelly WK, Trabulsi EJ, Lallas CD, Gomella LG, Knudsen KE. Novel actions of next-generation taxanes benefit advanced stages of prostate cancer. Clin Cancer Res 2015; 21:795-807. [PMID: 25691773 PMCID: PMC4333741 DOI: 10.1158/1078-0432.ccr-14-1358] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE To improve the outcomes of patients with castration-resistant prostate cancer (CRPC), there is an urgent need for more effective therapies and approaches that individualize specific treatments for patients with CRPC. These studies compared the novel taxane cabazitaxel with the previous generation docetaxel, and aimed to determine which tumors are most likely to respond. EXPERIMENTAL DESIGN Cabazitaxel and docetaxel were compared via in vitro modeling to determine the molecular mechanism, biochemical and cell biologic impact, and cell proliferation, which was further assessed ex vivo in human tumor explants. Isogenic pairs of RB knockdown and control cells were interrogated in vitro and in xenograft tumors for cabazitaxel response. RESULTS The data herein show that (i) cabazitaxel exerts stronger cytostatic and cytotoxic response compared with docetaxel, especially in CRPC; (ii) cabazitaxel induces aberrant mitosis, leading to pyknotic and multinucleated cells; (iii) taxanes do not act through the androgen receptor (AR); (iv) gene-expression profiling reveals distinct molecular actions for cabazitaxel; and (v) tumors that have progressed to castration resistance via loss of RB show enhanced sensitivity to cabazitaxel. CONCLUSIONS Cabazitaxel not only induces improved cytostatic and cytotoxic effects, but also affects distinct molecular pathways, compared with docetaxel, which could underlie its efficacy after docetaxel treatment has failed in patients with CRPC. Finally, RB is identified as the first potential biomarker that could define the therapeutic response to taxanes in metastatic CRPC. This would suggest that loss of RB function induces sensitization to taxanes, which could benefit up to 50% of CRPC cases.
Collapse
Affiliation(s)
- Renée de Leeuw
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Lisa D Berman-Booty
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Matthew J Schiewer
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Stephen J Ciment
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Robert B Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania. Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam P Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania. Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - William K Kelly
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Edouard J Trabulsi
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Costas D Lallas
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Leonard G Gomella
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania. Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
159
|
Sun T, Ye H, Wu CL, Lee GSM, Kantoff PW. Emerging players in prostate cancer: long non-coding RNAs. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2014; 2:294-299. [PMID: 25606575 PMCID: PMC4297325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/09/2014] [Indexed: 06/04/2023]
Abstract
Recent observations of novel long non-coding RNAs (lncRNAs) have considerably altered our understanding of cell biology. The role of lncRNAs as tumor suppressors or oncogenes has been extensively studied. Over-expression of oncogenic lncRNAs promotes tumor-cell proliferation and metastasis through chromatin looping and distal engagement with the androgen receptor, anti-sense gene regulation, alternative splicing, and impeding DNA repair. Prostate cancer is the most common type of cancer and frequent cause of cancer-related mortality in men worldwide. Unraveling the molecular and biological processes that contribute to prostate cancer development and progression is a challenging task. In prostate cancer, aberrant expression of lncRNAs has been associated with disease progression. In this review, we highlight the emerging impact of lncRNAs in prostate cancer research, with a particular focus on the mechanisms and functions of lncRNAs. Increased research on lncRNAs will lead to a greater understanding of prostate cancercinogenesis and progression and may lead to novel clinical applications. LncRNAs have great potential to become new biomarkers for detection, prognostication and prediction in prostate cancer.
Collapse
Affiliation(s)
- Tong Sun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical SchoolUSA
| | - Huihui Ye
- Department of Pathology, Beth-Israel Deaconess Medical Center, Harvard Medical SchoolUSA
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical SchoolUSA
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical SchoolUSA
| | - Philip W Kantoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical SchoolUSA
| |
Collapse
|
160
|
Shackney SE, Chowdhury SA, Schwartz R. A Novel Subset of Human Tumors That Simultaneously Overexpress Multiple E2F-responsive Genes Found in Breast, Ovarian, and Prostate Cancers. Cancer Inform 2014; 13:89-100. [PMID: 25392696 PMCID: PMC4221091 DOI: 10.4137/cin.s14062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/08/2014] [Accepted: 09/15/2014] [Indexed: 01/14/2023] Open
Abstract
Reasoning that overexpression of multiple E2F-responsive genes might be a useful marker for RB1 dysfunction, we compiled a list of E2F-responsive genes from the literature and evaluated their expression in publicly available gene expression microarray data of patients with breast cancer, serous ovarian cancer, and prostate cancer. In breast cancer, a group of tumors was identified, each of which simultaneously overexpressed multiple E2F-responsive genes. Seventy percent of these genes were concerned with cell cycle progression, DNA repair, or mitosis. These E2F-responsive gene overexpressing (ERGO) tumors frequently exhibited additional evidence of Rb/E2F axis dysfunction, were mostly triple negative, and preferentially overexpressed multiple basal cytokeratins, suggesting that they overlapped substantially with the basal-like tumor subset. ERGO tumors were also identified in serous ovarian cancer and prostate cancer. In these cancer types, there was no evidence for a tumor subset comparable to the breast cancer basal-like subset. A core group of about 30 E2F-responsive genes were overexpressed in all three cancer types. Thus, it appears that disorders of the Rb/E2F axis can arise at multiple organ sites and produce tumors that simultaneously overexpress multiple E2F-responsive genes.
Collapse
Affiliation(s)
- Stanley E Shackney
- President and CEO, Intelligent Oncotherapeutics Incorporated, Pittsburgh, PA, USA
| | - Salim Akhter Chowdhury
- Graduate student of Computational Biology, Lane Center for Computational Biology and Joint Carnegie Mellon/University of Pittsburgh Ph.D. Program in Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Russell Schwartz
- Professor of Biological Sciences and Computational Biology, Department of Biological Sciences and Lane Center for Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
161
|
Increased expression of CYP17A1 indicates an effective targeting of the androgen receptor axis in castration resistant prostate cancer (CRPC). SPRINGERPLUS 2014; 3:574. [PMID: 25332874 PMCID: PMC4193971 DOI: 10.1186/2193-1801-3-574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/26/2014] [Indexed: 01/18/2023]
Abstract
Recent breakthrough therapies targeting androgen receptor signalling in castration resistant prostate cancer (CRPC) involve multifunctional androgen receptor (AR) blockade and exhaustive androgen deprivation. Nevertheless, limitations to an enduring effectiveness of new drugs are anticipated in resistance mechanisms occurring under such treatments. In this study we used CRPC cell models VCaP and LNCaP as well as AR-negative PC-3- and non-neoplastic epithelial BPH-1-cells treated with 5, 10 or 25 μmol/L abiraterone hydrolyzed from abiraterone acetate (AA). The origin of CYP17A1 up-regulation under AA treatment was investigated in CRPC cell models by qRT-PCR and western-blot procedures. AA treatments of AR positive CRPC cell models led to decreased expression of androgen regulated genes such as PSA. In these cells diminished expression of androgen regulated genes was accompanied by an up-regulation of CYP17A1 expression within short-term treatments. No such effects became evident in AR-negative PC-3 cells. AR directed siRNA (siAR) used in VCaP cells significantly reduced mRNA expression and AR protein abundance. Such interference with AR signalling in the absence of abiraterone acetate also caused a marked up-regulation of CYP17A1 expression. Down-regulation of androgen regulated genes occurs in spite of an elevated expression of CYP17A1, the very target enzyme for this drug. CYP17A1 up-regulation already takes place within such short treatments with AA and does not require adaptation events over several cell cycles. CYP17A1 is also up-regulated in the absence of AA when AR signalling is physically eliminated by siAR. These results reveal an immediate counter-regulation of CYP17A1 expression whenever AR-signalling is inhibited adequately but not a persisting adaptation yielding drug resistance.
Collapse
|
162
|
Toki H, Inoue M, Minowa O, Motegi H, Saiki Y, Wakana S, Masuya H, Gondo Y, Shiroishi T, Yao R, Noda T. Novel retinoblastoma mutation abrogating the interaction to E2F2/3, but not E2F1, led to selective suppression of thyroid tumors. Cancer Sci 2014; 105:1360-8. [PMID: 25088905 PMCID: PMC4462357 DOI: 10.1111/cas.12495] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/18/2014] [Accepted: 07/25/2014] [Indexed: 01/18/2023] Open
Abstract
Mutant mouse models are indispensable tools for clarifying gene functions and elucidating the pathogenic mechanisms of human diseases. Here, we describe novel cancer models bearing point mutations in the retinoblastoma gene (Rb1) generated by N-ethyl-N-nitrosourea mutagenesis. Two mutations in splice sites reduced Rb1 expression and led to a tumor spectrum and incidence similar to those observed in the conventional Rb1 knockout mice. The missense mutant, Rb1D326V/+, developed pituitary tumors, but thyroid tumors were completely suppressed. Immunohistochemical analyses of thyroid tissue revealed that E2F1, but not E2F2/3, was selectively inactivated, indicating that the mutant Rb protein (pRb) suppressed thyroid tumors by inactivating E2F1. Interestingly, Rb1D326V/+ mice developed pituitary tumors that originated from the intermediate lobe of the pituitary, despite selective inactivation of E2F1. Furthermore, in the anterior lobe of the pituitary, other E2F were also inactivated. These observations show that pRb mediates the inactivation of E2F function and its contribution to tumorigenesis is highly dependent on the cell type. Last, by using a reconstitution assay of synthesized proteins, we showed that the D326V missense pRb bound to E2F1 but failed to interact with E2F2/3. These results reveal the effect of the pRb N-terminal domain on E2F function and the impact of the protein on tumorigenesis. Thus, this mutant mouse model can be used to investigate human Rb family-bearing mutations at the N-terminal region.
Collapse
Affiliation(s)
- Hideaki Toki
- Team for Advanced Development and Evaluation of Human Disease Models, Riken BioResource Center, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Shtivelman E, Beer TM, Evans CP. Molecular pathways and targets in prostate cancer. Oncotarget 2014; 5:7217-59. [PMID: 25277175 PMCID: PMC4202120 DOI: 10.18632/oncotarget.2406] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/28/2014] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer co-opts a unique set of cellular pathways in its initiation and progression. The heterogeneity of prostate cancers is evident at earlier stages, and has led to rigorous efforts to stratify the localized prostate cancers, so that progression to advanced stages could be predicted based upon salient features of the early disease. The deregulated androgen receptor signaling is undeniably most important in the progression of the majority of prostate tumors. It is perhaps because of the primacy of the androgen receptor governed transcriptional program in prostate epithelium cells that once this program is corrupted, the consequences of the ensuing changes in activity are pleotropic and could contribute to malignancy in multiple ways. Following localized surgical and radiation therapies, 20-40% of patients will relapse and progress, and will be treated with androgen deprivation therapies. The successful development of the new agents that inhibit androgen signaling has changed the progression free survival in hormone resistant disease, but this has not changed the almost ubiquitous development of truly resistant phenotypes in advanced prostate cancer. This review summarizes the current understanding of the molecular pathways involved in localized and metastatic prostate cancer, with an emphasis on the clinical implications of the new knowledge.
Collapse
Affiliation(s)
| | - Tomasz M. Beer
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Christopher P. Evans
- Department of Urology and Comprehensive Cancer Center, University of California Davis, Davis, CA
| |
Collapse
|
164
|
Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, Dowling C, Wanjala JN, Undvall EA, Arora VK, Wongvipat J, Kossai M, Ramazanoglu S, Barboza LP, Di W, Cao Z, Zhang QF, Sirota I, Ran L, MacDonald TY, Beltran H, Mosquera JM, Touijer KA, Scardino PT, Laudone VP, Curtis KR, Rathkopf DE, Morris MJ, Danila DC, Slovin SF, Solomon SB, Eastham JA, Chi P, Carver B, Rubin MA, Scher HI, Clevers H, Sawyers CL, Chen Y. Organoid cultures derived from patients with advanced prostate cancer. Cell 2014; 159:176-187. [PMID: 25201530 DOI: 10.1016/j.cell.2014.08.016] [Citation(s) in RCA: 1075] [Impact Index Per Article: 107.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/22/2014] [Accepted: 08/12/2014] [Indexed: 12/11/2022]
Abstract
The lack of in vitro prostate cancer models that recapitulate the diversity of human prostate cancer has hampered progress in understanding disease pathogenesis and therapy response. Using a 3D organoid system, we report success in long-term culture of prostate cancer from biopsy specimens and circulating tumor cells. The first seven fully characterized organoid lines recapitulate the molecular diversity of prostate cancer subtypes, including TMPRSS2-ERG fusion, SPOP mutation, SPINK1 overexpression, and CHD1 loss. Whole-exome sequencing shows a low mutational burden, consistent with genomics studies, but with mutations in FOXA1 and PIK3R1, as well as in DNA repair and chromatin modifier pathways that have been reported in advanced disease. Loss of p53 and RB tumor suppressor pathway function are the most common feature shared across the organoid lines. The methodology described here should enable the generation of a large repertoire of patient-derived prostate cancer lines amenable to genetic and pharmacologic studies.
Collapse
Affiliation(s)
- Dong Gao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ian Vela
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Sboner
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA; Institute for Precision Medicine of Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Phillip J Iaquinta
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wouter R Karthaus
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
| | - Anuradha Gopalan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Catherine Dowling
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jackline N Wanjala
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eva A Undvall
- Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vivek K Arora
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - John Wongvipat
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Myriam Kossai
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Sinan Ramazanoglu
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Luendreo P Barboza
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wei Di
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zhen Cao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Qi Fan Zhang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Inna Sirota
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Leili Ran
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Theresa Y MacDonald
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Himisha Beltran
- Institute for Precision Medicine of Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Juan-Miguel Mosquera
- Institute for Precision Medicine of Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Karim A Touijer
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Peter T Scardino
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vincent P Laudone
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kristen R Curtis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dana E Rathkopf
- Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael J Morris
- Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daniel C Danila
- Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Susan F Slovin
- Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen B Solomon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - James A Eastham
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Brett Carver
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mark A Rubin
- Institute for Precision Medicine of Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Howard I Scher
- Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
| | - Charles L Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
165
|
Meier C, Spitschak A, Abshagen K, Gupta S, Mor JM, Wolkenhauer O, Haier J, Vollmar B, Alla V, Pützer BM. Association of RHAMM with E2F1 promotes tumour cell extravasation by transcriptional up-regulation of fibronectin. J Pathol 2014; 234:351-64. [PMID: 25042645 DOI: 10.1002/path.4400] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/23/2014] [Accepted: 06/26/2014] [Indexed: 01/04/2023]
Abstract
Dissemination of cancer cells from primary to distant sites is a complex process; little is known about the genesis of metastatic changes during disease development. Here we show that the metastatic potential of E2F1-dependent circulating tumour cells (CTCs) relies on a novel function of the hyaluronan-mediated motility receptor RHAMM. E2F1 directly up-regulates RHAMM, which in turn acts as a co-activator of E2F1 to stimulate expression of the extracellular matrix protein fibronectin. Enhanced fibronectin secretion links E2F1/RHAMM transcriptional activity to integrin-β1-FAK signalling associated with cytoskeletal remodelling and enhanced tumour cell motility. RHAMM depletion abolishes fibronectin expression and cell transmigration across the endothelial layer in E2F1-activated cells. In a xenograft model, knock-down of E2F1 or RHAMM in metastatic cells protects the liver parenchyma of mice against extravasation of CTCs, whereas the number of transmigrated cells increases in response to E2F1 induction. Expression data from clinical tissue samples reveals high E2F1 and RHAMM levels that closely correlate with malignant progression. These findings suggest a requirement for RHAMM in late-stage metastasis by a mechanism involving cooperative stimulation of fibronectin, with a resultant tumourigenic microenvironment important for enhanced extravasation and distant organ colonization. Therefore, stimulation of the E2F1-RHAMM axis in aggressive cancer cells is of high clinical significance. Targeting RHAMM may represent a promising approach to avoid E2F1-mediated metastatic dissemination.
Collapse
Affiliation(s)
- Claudia Meier
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Centre, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Thangavel C, Boopathi E, Ciment S, Liu Y, O'Neill R, Sharma A, McMahon SB, Mellert H, Addya S, Ertel A, Birbe R, Fortina P, Dicker AP, Knudsen KE, Den RB. The retinoblastoma tumor suppressor modulates DNA repair and radioresponsiveness. Clin Cancer Res 2014; 20:5468-5482. [PMID: 25165096 DOI: 10.1158/1078-0432.ccr-14-0326] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Perturbations in the retinoblastoma pathway are over-represented in advanced prostate cancer; retinoblastoma loss promotes bypass of first-line hormone therapy. Conversely, preliminary studies suggested that retinoblastoma-deficient tumors may become sensitized to a subset of DNA-damaging agents. Here, the molecular and in vivo consequence of retinoblastoma status was analyzed in models of clinical relevance. EXPERIMENTAL DESIGN Experimental work was performed with multiple isogenic prostate cancer cell lines (hormone sensitive: LNCaP and LAPC4 cells and hormone resistant C42, 22Rv1 cells; stable knockdown of retinoblastoma using shRNA). Multiple mechanisms were interrogated including cell cycle, apoptosis, and DNA damage repair. Transcriptome analysis was performed, validated, and mechanisms discerned. Cell survival was measured using clonogenic cell survival assay and in vivo analysis was performed in nude mice with human derived tumor xenografts. RESULTS Loss of retinoblastoma enhanced the radioresponsiveness of both hormone-sensitive and castrate-resistant prostate cancer. Hypersensitivity to ionizing radiation was not mediated by cell cycle or p53. Retinoblastoma loss led to alteration in DNA damage repair and activation of the NF-κB pathway and subsequent cellular apoptosis through PLK3. In vivo xenografts of retinoblastoma-deficient tumors exhibited diminished tumor mass, lower PSA kinetics, and decreased tumor growth after treatment with ionizing radiation (P < 0.05). CONCLUSIONS Loss of retinoblastoma confers increased radiosensitivity in prostate cancer. This hypersensitization was mediated by alterations in apoptotic signaling. Combined, these not only provide insight into the molecular consequence of retinoblastoma loss, but also credential retinoblastoma status as a putative biomarker for predicting response to radiotherapy.
Collapse
Affiliation(s)
| | - Ettickan Boopathi
- Department of Surgery, Division of Urology, Glenolden, Pennsylvania, USA
| | - Steve Ciment
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Yi Liu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Raymond O'Neill
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ankur Sharma
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steve B McMahon
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Hestia Mellert
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Biomedical Graduate Studies, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Colorado, USA
| | - Sankar Addya
- Cancer Genomics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam Ertel
- Cancer Genomics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ruth Birbe
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Paolo Fortina
- Cancer Genomics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam P Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Karen E Knudsen
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert B Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
167
|
Gu Y, Cheng Y, Song Y, Zhang Z, Deng M, Wang C, Zheng G, He Z. MicroRNA-493 suppresses tumor growth, invasion and metastasis of lung cancer by regulating E2F1. PLoS One 2014; 9:e102602. [PMID: 25105419 PMCID: PMC4126682 DOI: 10.1371/journal.pone.0102602] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023] Open
Abstract
miRNAs have been proposed to be key regulators of progression and metastasis in cancer. However, an understanding of their roles and molecular mechanisms is needed to provide deeper insights for better therapeutic opportunities. In this study we investigated the role and mechanism of miR-493 in the development and progression of nonsmall-cell lung cancer (NSCLC). Our data indicated that the expression of miR-493 was markedly reduced in pulmonary carcinoma. The ectopic expression of miR-493 impaired cell growth and invasion in vitro and in vivo. Mechanically, miR-493 commonly directly targeted E2F1, which resulted in a robust reduction of the expression of mRNA and protein. This effect, in turn, decreased the growth, invasion and metastasis of lung cancer cells. Our findings highlight the importance of miR-493 dysfunction in promoting tumor progression, and implicate miR-493 as a potential therapeutic target in lung cancer.
Collapse
Affiliation(s)
- Yixue Gu
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
- Medical School, University of South China, Hengyang, Hunan, PR China
| | - Ye Cheng
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Ying Song
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Zhijie Zhang
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Min Deng
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chengkun Wang
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Guopei Zheng
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Zhimin He
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
- * E-mail:
| |
Collapse
|
168
|
Ware KE, Garcia-Blanco MA, Armstrong AJ, Dehm SM. Biologic and clinical significance of androgen receptor variants in castration resistant prostate cancer. Endocr Relat Cancer 2014; 21:T87-T103. [PMID: 24859991 PMCID: PMC4277180 DOI: 10.1530/erc-13-0470] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As prostate cancer (PCa) progresses to the lethal castration resistant and metastatic form, genetic and epigenetic adaptation, clonal selection, and evolution of the tumor microenvironment contribute to the emergence of unique biological characteristics under the selective pressure of external stresses. These stresses include the therapies applied in the clinic or laboratory and the exposures of cancers to hormonal, paracrine, or autocrine stimuli in the context of the tumor micro- and macro-environment. The androgen receptor (AR) is a key gene involved in PCa etiology and oncogenesis, including disease development, progression, response to initial hormonal therapies, and subsequent resistance to hormonal therapies. Alterations in the AR signaling pathway have been observed in certain selection contexts and contribute to the resistance to agents that target hormonal regulation of the AR, including standard androgen deprivation therapy, antiandrogens such as enzalutamide, and androgen synthesis inhibition with abiraterone acetate. One such resistance mechanism is the synthesis of constitutively active AR variants lacking the canonical ligand-binding domain. This review focuses on the etiology, characterization, biological properties, and emerging data contributing to the clinical characteristics of AR variants, and suggests approaches to full-length AR and AR variant biomarker validation, assessment, and systemic targeting in the clinic.
Collapse
Affiliation(s)
- Kathryn E Ware
- Departments of Molecular Genetics and MedicineDuke University, 213 Research Dr, 0045 CARL Building, Durham, North Carolina 27710, USADepartment of MedicineDuke Cancer Institute, Duke University, Durham, North Carolina, USAMasonic Cancer CenterUniversity of Minnesota Masonic Cancer Center, Mayo Mail Code 806, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USADepartment of Laboratory Medicine and PathologyUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Mariano A Garcia-Blanco
- Departments of Molecular Genetics and MedicineDuke University, 213 Research Dr, 0045 CARL Building, Durham, North Carolina 27710, USADepartment of MedicineDuke Cancer Institute, Duke University, Durham, North Carolina, USAMasonic Cancer CenterUniversity of Minnesota Masonic Cancer Center, Mayo Mail Code 806, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USADepartment of Laboratory Medicine and PathologyUniversity of Minnesota, Minneapolis, Minnesota, USADepartments of Molecular Genetics and MedicineDuke University, 213 Research Dr, 0045 CARL Building, Durham, North Carolina 27710, USADepartment of MedicineDuke Cancer Institute, Duke University, Durham, North Carolina, USAMasonic Cancer CenterUniversity of Minnesota Masonic Cancer Center, Mayo Mail Code 806, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USADepartment of Laboratory Medicine and PathologyUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew J Armstrong
- Departments of Molecular Genetics and MedicineDuke University, 213 Research Dr, 0045 CARL Building, Durham, North Carolina 27710, USADepartment of MedicineDuke Cancer Institute, Duke University, Durham, North Carolina, USAMasonic Cancer CenterUniversity of Minnesota Masonic Cancer Center, Mayo Mail Code 806, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USADepartment of Laboratory Medicine and PathologyUniversity of Minnesota, Minneapolis, Minnesota, USADepartments of Molecular Genetics and MedicineDuke University, 213 Research Dr, 0045 CARL Building, Durham, North Carolina 27710, USADepartment of MedicineDuke Cancer Institute, Duke University, Durham, North Carolina, USAMasonic Cancer CenterUniversity of Minnesota Masonic Cancer Center, Mayo Mail Code 806, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USADepartment of Laboratory Medicine and PathologyUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Scott M Dehm
- Departments of Molecular Genetics and MedicineDuke University, 213 Research Dr, 0045 CARL Building, Durham, North Carolina 27710, USADepartment of MedicineDuke Cancer Institute, Duke University, Durham, North Carolina, USAMasonic Cancer CenterUniversity of Minnesota Masonic Cancer Center, Mayo Mail Code 806, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USADepartment of Laboratory Medicine and PathologyUniversity of Minnesota, Minneapolis, Minnesota, USADepartments of Molecular Genetics and MedicineDuke University, 213 Research Dr, 0045 CARL Building, Durham, North Carolina 27710, USADepartment of MedicineDuke Cancer Institute, Duke University, Durham, North Carolina, USAMasonic Cancer CenterUniversity of Minnesota Masonic Cancer Center, Mayo Mail Code 806, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USADepartment of Laboratory Medicine and PathologyUniversity of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
169
|
Muniyan S, Ingersoll MA, Batra SK, Lin MF. Cellular prostatic acid phosphatase, a PTEN-functional homologue in prostate epithelia, functions as a prostate-specific tumor suppressor. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1846:88-98. [PMID: 24747769 PMCID: PMC4140952 DOI: 10.1016/j.bbcan.2014.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 04/11/2014] [Indexed: 12/13/2022]
Abstract
The inactivation of tumor suppressor genes (TSGs) plays a vital role in the progression of human cancers. Nevertheless, those ubiquitous TSGs have been shown with limited roles in various stages of diverse carcinogenesis. Investigation on identifying unique TSG, especially for early stage of carcinogenesis, is imperative. As such, the search for organ-specific TSGs has emerged as a major strategy in cancer research. Prostate cancer (PCa) has the highest incidence in solid tumors in US males. Cellular prostatic acid phosphatase (cPAcP) is a prostate-specific differentiation antigen. Despite intensive studies over the past several decades on PAcP as a PCa biomarker, the role of cPAcP as a PCa-specific tumor suppressor has only recently been emerged and validated. The mechanism underlying the pivotal role of cPAcP as a prostate-specific TSG is, in part, due to its function as a protein tyrosine phosphatase (PTP) as well as a phosphoinositide phosphatase (PIP), an apparent functional homologue to phosphatase and tensin homolog (PTEN) in PCa cells. This review is focused on discussing the function of this authentic prostate-specific tumor suppressor and the mechanism behind the loss of cPAcP expression leading to prostate carcinogenesis. We review other phosphatases' roles as TSGs which regulate oncogenic PI3K signaling in PCa and discuss the functional similarity between cPAcP and PTEN in prostate carcinogenesis.
Collapse
Affiliation(s)
- Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew A Ingersoll
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Department of Surgery/Urology, University of Nebraska Medical Center, Omaha, NE, USA; College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC.
| |
Collapse
|
170
|
Itsumi M, Shiota M, Yokomizo A, Takeuchi A, Kashiwagi E, Dejima T, Inokuchi J, Tatsugami K, Uchiumi T, Naito S. PMA induces androgen receptor downregulation and cellular apoptosis in prostate cancer cells. J Mol Endocrinol 2014; 53:31-41. [PMID: 24780839 DOI: 10.1530/jme-13-0303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Phorbol 12-myristate 13-acetate (PMA) induces cellular apoptosis in prostate cancer cells, the growth of which is governed by androgen/androgen receptor (AR) signaling, but the mechanism by which PMA exerts this effect remains unknown. Therefore, in this study, we investigated the mechanistic action of PMA in prostate cancer cells with regard to AR. We showed that PMA decreased E2F1 as well as AR expression in androgen-dependent prostate cancer LNCaP cells. Furthermore, PMA activated JNK and p53 signaling, resulting in the induction of cellular apoptosis. In LNCaP cells, androgen deprivation and a novel anti-androgen enzalutamide (MDV3100) augmented cellular apoptosis induced by PMA. Moreover, castration-resistant prostate cancer (CRPC) C4-2 cells were more sensitive to PMA compared with LNCaP cells and were sensitized to PMA by enzalutamide. Finally, the expression of PKC, E2F1, and AR was diminished in PMA-resistant cells, indicating that the gain of independence from PKC, E2F1, and AR functions leads to PMA resistance. In conclusion, PMA exerted its anti-cancer effects via the activation of pro-apoptotic JNK/p53 and inhibition of pro-proliferative E2F1/AR in prostate cancer cells including CRPC cells. The therapeutic effects of PMA were augmented by androgen deletion and enzalutamide in androgen-dependent prostate cancer cells, as well as by enzalutamide in castration-resistant cells. Taken together, PMA derivatives may be promising therapeutic agents for treating prostate cancer patients including CRPC patients.
Collapse
Affiliation(s)
- Momoe Itsumi
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaki Shiota
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akira Yokomizo
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ario Takeuchi
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Eiji Kashiwagi
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Dejima
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Junichi Inokuchi
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Katsunori Tatsugami
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Uchiumi
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Seiji Naito
- Departments of UrologyClinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
171
|
Georgi B, Korzeniewski N, Hadaschik B, Grüllich C, Roth W, Sültmann H, Pahernik S, Hohenfellner M, Duensing S. Evolving therapeutic concepts in prostate cancer based on genome-wide analyses (review). Int J Oncol 2014; 45:1337-44. [PMID: 25070358 DOI: 10.3892/ijo.2014.2567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/09/2014] [Indexed: 11/05/2022] Open
Abstract
Treatment of castration resistant prostate cancer (CRPC) continues to represent a major urooncological challenge due to tumor heterogeneity and the inevitable development of therapy resistance. Although androgen deprivation therapy retains an important role in the management of CRPC, recent evidence suggests that a broader spectrum of therapeutic targets may improve patient response and delay development of advanced disease. Genome-wide analyses have identified four major signaling nodes that are most frequently altered in prostate cancer: i) the androgen receptor (AR); ii) the PI3K pathway; iii) the Ras/Raf/MEK/ERK pathway; and iv) the retinoblastoma protein (pRB) signaling pathway. Extensive crosstalk and redundancy exists between these signaling pathways, which underscores the need for combination therapies. There are several novel AR pathway inhibitors currently in clinical use. Clinical trials are being performed on single-agent PI3K inhibitors with some success in tumors with genetically altered PI3K components. MEK/ERK inhibitors are also in clinical trials and the importance of pRB inactivation in prostate cancer is becoming more widely recognized. A greater understanding of the effects of single agent therapy on compensatory signaling pathway activation that can potentially thwart antitumoral responses is urgently needed and will provide additional insight into the mechanism of therapy resistance and how to further delay the progression to lethal disease.
Collapse
Affiliation(s)
- Björn Georgi
- Department of Urology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| | - Nina Korzeniewski
- Section of Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| | - Boris Hadaschik
- Department of Urology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| | - Carsten Grüllich
- National Center for Tumor Diseases, Department of Medical Oncology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| | - Wilfried Roth
- Department of Pathology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| | - Holger Sültmann
- Division for Cancer Genome Research, National Center for Tumor Diseases and German Cancer Research Center, D-69120 Heidelberg, Germany
| | - Sascha Pahernik
- Department of Urology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, University of Heidelberg School of Medicine, D-69120 Heidelberg, Germany
| |
Collapse
|
172
|
Esch L, Schulz WA, Albers P. Sequential Treatment with Taxanes and Novel Anti-Androgenic Compounds in Castration-Resistant Prostate Cancer. Oncol Res Treat 2014; 37:492-8. [DOI: 10.1159/000365530] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 06/27/2014] [Indexed: 11/19/2022]
|
173
|
A TRIP230-retinoblastoma protein complex regulates hypoxia-inducible factor-1α-mediated transcription and cancer cell invasion. PLoS One 2014; 9:e99214. [PMID: 24919196 PMCID: PMC4053355 DOI: 10.1371/journal.pone.0099214] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/12/2014] [Indexed: 12/15/2022] Open
Abstract
Localized hypoxia in solid tumors activates transcriptional programs that promote the metastatic transformation of cells. Like hypoxia-inducible hyper-vascularization, loss of the retinoblastoma protein (Rb) is a trait common to advanced stages of tumor progression in many metastatic cancers. However, no link between the role of Rb and hypoxia-driven metastatic processes has been established. We demonstrated that Rb is a key mediator of the hypoxic response mediated by HIF1α/β, the master regulator of the hypoxia response, and its essential co-activator, the thyroid hormone receptor/retinoblastoma-interacting protein (TRIP230). Furthermore, loss of Rb unmasks the full co-activation potential of TRIP230. Using small inhibitory RNA approaches in vivo, we established that Rb attenuates the normal physiological response to hypoxia by HIF1α. Notably, loss of Rb results in hypoxia-dependent biochemical changes that promote acquisition of an invasive phenotype in MCF7 breast cancer cells. In addition, Rb is present in HIF1α-ARNT/HIF1β transcriptional complexes associated with TRIP230 as determined by co-immuno-precipitation, GST-pull-down and ChIP assays. These results demonstrate that Rb is a negative modulator of hypoxia-regulated transcription by virtue of its direct effects on the HIF1 complex. This work represents the first link between the functional ablation of Rb in tumor cells and HIF1α-dependent transcriptional activation and invasion.
Collapse
|
174
|
Rico-Bautista E, Zhu W, Kitada S, Ganapathy S, Lau E, Krajewski S, Ramirez J, Bush JA, Yuan Z, Wolf DA. Small molecule-induced mitochondrial disruption directs prostate cancer inhibition via UPR signaling. Oncotarget 2014; 4:1212-29. [PMID: 23902736 PMCID: PMC3787152 DOI: 10.18632/oncotarget.1130] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We previously identified SMIP004 (N-(4-butyl-2-methyl-phenyl) acetamide) as a novel inducer of cancer-cell selective apoptosis of human prostate cancer cells. SMIP004 decreased the levels of positive cell cycle regulators, upregulated cyclin-dependent kinase inhibitors, and resulted in G1 arrest, inhibition of colony formation in soft agar, and cell death. However, the mechanism of SMIP004-induced cancer cell selective apoptosis remained unknown. Here, we used chemical genomic and proteomic profiling to unravel a SMIP004-induced pro-apoptotic pathway, which initiates with disruption of mitochondrial respiration leading to oxidative stress. This, in turn, activates two pathways, one eliciting cell cycle arrest by rapidly targeting cyclin D1 for proteasomal degradation and driving the transcriptional downregulation of the androgen receptor, and a second pathway that activates pro-apoptotic signaling through MAPK activation downstream of the unfolded protein response (UPR). SMIP004 potently inhibits the growth of prostate and breast cancer xenografts in mice. Our data suggest that SMIP004, by inducing mitochondrial ROS formation, targets specific sensitivities of prostate cancer cells to redox and bioenergetic imbalances that can be exploited in cancer therapy.
Collapse
|
175
|
Yuan X, Cai C, Chen S, Chen S, Yu Z, Balk SP. Androgen receptor functions in castration-resistant prostate cancer and mechanisms of resistance to new agents targeting the androgen axis. Oncogene 2014; 33:2815-25. [PMID: 23752196 PMCID: PMC4890635 DOI: 10.1038/onc.2013.235] [Citation(s) in RCA: 261] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 04/30/2013] [Accepted: 05/06/2013] [Indexed: 12/17/2022]
Abstract
The metabolic functions of androgen receptor (AR) in normal prostate are circumvented in prostate cancer (PCa) to drive tumor growth, and the AR also can acquire new growth-promoting functions during PCa development and progression through genetic and epigenetic mechanisms. Androgen deprivation therapy (ADT, surgical or medical castration) is the standard treatment for metastatic PCa, but patients invariably relapse despite castrate androgen levels (castration-resistant PCa, CRPC). Early studies from many groups had shown that AR was highly expressed and transcriptionally active in CRPC, and indicated that steroids from the adrenal glands were contributing to this AR activity. More recent studies showed that CRPC cells had increased expression of enzymes mediating androgen synthesis from adrenal steroids, and could synthesize androgens de novo from cholesterol. Phase III clinical trials showing a survival advantage in CRPC for treatment with abiraterone (inhibitor of the enzyme CYP17A1 required for androgen synthesis that markedly reduces androgens and precursor steroids) and for enzalutamide (new AR antagonist) have now confirmed that AR activity driven by residual androgens makes a major contribution to CRPC, and led to the recent Food and Drug Administration approval of both agents. Unfortunately, patients treated with these agents for advanced CRPC generally relapse within a year and AR appears to be active in the relapsed tumors, but the molecular mechanisms mediating intrinsic or acquired resistance to these AR-targeted therapies remain to be defined. This review outlines AR functions that contribute to PCa development and progression, the roles of intratumoral androgen synthesis and AR structural alterations in driving AR activity in CRPC, mechanisms of action for abiraterone and enzalutamide, and possible mechanisms of resistance to these agents.
Collapse
MESH Headings
- Androgen Receptor Antagonists/therapeutic use
- Androgens/metabolism
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Disease Progression
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Receptors, Androgen/chemistry
- Receptors, Androgen/metabolism
- Repressor Proteins/metabolism
- Steroid 17-alpha-Hydroxylase/antagonists & inhibitors
- Steroid 17-alpha-Hydroxylase/metabolism
- Trans-Activators/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- X Yuan
- Hematology Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - C Cai
- Hematology Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - S Chen
- Hematology Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - S Chen
- Hematology Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Z Yu
- Hematology Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - S P Balk
- Hematology Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
176
|
The combination of the prodrugs perforin-CEBPD and perforin-granzyme B efficiently enhances the activation of caspase signaling and kills prostate cancer. Cell Death Dis 2014; 5:e1220. [PMID: 24810056 PMCID: PMC4047860 DOI: 10.1038/cddis.2014.106] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/11/2014] [Accepted: 02/11/2014] [Indexed: 11/09/2022]
Abstract
The survival of prostate cancer (PrCa) patients is associated with the transition to hormone-independent tumor growth and metastasis. Clinically, the dysregulation of androgen action has been associated with the formation of PrCa and the outcome of androgen deprivation therapy in PrCa. CCAAT/enhancer binding protein delta (CEBPD) is a transcription factor that has been reported to act as an oncogene or tumor suppressor, depending on the extra- and intracellular environments following tumorigenesis. We found that androgen can activate CEBPD transcription by direct binding of the androgen receptor (AR) to the CEBPD promoter region. Increases of suppressor of zeste 12 (SUZ12) and enhancer of zeste homolog 2 (EZH2) attenuated the androgen-induced transcription of CEBPD. Importantly, the increases in E2F1, SUZ12 and EZH2 as well as the inactivation of CEBPD were associated with the clinicopathological variables and survival of PrCa patients. We revealed that caspase 8 (CASP8), an apoptotic initiator, is responsive to CEBPD induction. Reporter and in vivo DNA-binding assays revealed that CEBPD directly binds to and activates CASP8 reporter activity. A prodrug system was developed for therapeutic application in AR-independent or androgen-insensitive PrCa to avoid the epigenetic effects on the suppression of CEBPD expression. Our results showed that the combination of a perforin (PF)-CEBPD prodrug (which increases the level of procaspase-8) and a PF-granzyme B prodrug (which activates CASP8 and caspase 3 (CASP3)) showed an additive effect in triggering the apoptotic pathway and enhancing apoptosis in PrCa cells.
Collapse
|
177
|
Ramos-Montoya A, Lamb AD, Russell R, Carroll T, Jurmeister S, Galeano-Dalmau N, Massie CE, Boren J, Bon H, Theodorou V, Vias M, Shaw GL, Sharma NL, Ross-Adams H, Scott HE, Vowler SL, Howat WJ, Warren AY, Wooster RF, Mills IG, Neal DE. HES6 drives a critical AR transcriptional programme to induce castration-resistant prostate cancer through activation of an E2F1-mediated cell cycle network. EMBO Mol Med 2014; 6:651-61. [PMID: 24737870 PMCID: PMC4023887 DOI: 10.1002/emmm.201303581] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Castrate-resistant prostate cancer (CRPC) is poorly characterized and heterogeneous and while the androgen receptor (AR) is of singular importance, other factors such as c-Myc and the E2F family also play a role in later stage disease. HES6 is a transcription co-factor associated with stem cell characteristics in neural tissue. Here we show that HES6 is up-regulated in aggressive human prostate cancer and drives castration-resistant tumour growth in the absence of ligand binding by enhancing the transcriptional activity of the AR, which is preferentially directed to a regulatory network enriched for transcription factors such as E2F1. In the clinical setting, we have uncovered a HES6-associated signature that predicts poor outcome in prostate cancer, which can be pharmacologically targeted by inhibition of PLK1 with restoration of sensitivity to castration. We have therefore shown for the first time the critical role of HES6 in the development of CRPC and identified its potential in patient-specific therapeutic strategies.
Collapse
Affiliation(s)
- Antonio Ramos-Montoya
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Alastair D Lamb
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK,*Corresponding author. Tel: +44 1223 331940; Fax: +44 1223 769007; E-mail:
| | - Roslin Russell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Thomas Carroll
- Bioinformatics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Sarah Jurmeister
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Nuria Galeano-Dalmau
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Charlie E Massie
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Joan Boren
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Helene Bon
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Vasiliki Theodorou
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Maria Vias
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Greg L Shaw
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK
| | - Naomi L Sharma
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK
| | - Helen Ross-Adams
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Helen E Scott
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Sarah L Vowler
- Bioinformatics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - William J Howat
- Histopathology/ISH Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Anne Y Warren
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Pathology, Addenbrooke's HospitalCambridge, UK
| | | | - Ian G Mills
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Prostate Cancer Research Group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of OsloOslo, Norway,Departments of Cancer Prevention and Urology, Institute of Cancer Research and Oslo University HospitalsOslo, Norway
| | - David E Neal
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK,Department of Urology, Addenbrooke's HospitalCambridge, UK,Department of Oncology, University of CambridgeCambridge, UK,**Corresponding author. Tel: +44 1223 331940; Fax: +44 1223 769007; E-mail:
| |
Collapse
|
178
|
Deletion of the olfactomedin 4 gene is associated with progression of human prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 183:1329-38. [PMID: 24070418 DOI: 10.1016/j.ajpath.2013.06.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/10/2013] [Accepted: 06/24/2013] [Indexed: 02/07/2023]
Abstract
The olfactomedin 4 (OLFM4) gene is located on chromosome 13q14.3, which frequently is deleted in human prostate cancer. However, direct genetic evidence of OLFM4 gene alteration in human prostate cancer has not yet been obtained. In this study, we investigated the genetics, protein expression, and functions of the OLFM4 gene in human prostate cancer. We found overall 25% deletions within the OLFM4 gene in cancerous epithelial cells compared with adjacent normal epithelial cells that were microdissected from 31 prostate cancer specimens using laser-capture microdissection and genomic DNA sequencing. We found 28% to 45% hemizygous and 15% to 57% homozygous deletions of the OLFM4 gene via fluorescence in situ hybridization analysis from 44 different prostate cancer patient samples. Moreover, homozygous deletion of the OLFM4 gene significantly correlated with advanced prostate cancer. By using immunohistochemical analysis of 162 prostate cancer tissue array samples representing a range of Gleason scores, we found that OLFM4 protein expression correlated inversely with advanced prostate cancer, consistent with the genetic results. We also showed that a truncated mutant of OLFM4 that lacks the olfactomedin domain eliminated suppression of PC-3 prostate cancer cell growth. Together, our findings indicate that OLFM4 is a novel candidate tumor-suppressor gene for chromosome 13q and may shed new light on strategies that could be used for the diagnosis, prognosis, and treatment of prostate cancer patients.
Collapse
|
179
|
Liu C, Liu A, Hu J, Yuan V, Halabi S. Adjusting for misclassification in a stratified biomarker clinical trial. Stat Med 2014; 33:3100-13. [PMID: 24733510 DOI: 10.1002/sim.6164] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 12/02/2013] [Accepted: 03/07/2014] [Indexed: 01/01/2023]
Abstract
Clinical trials utilizing predictive biomarkers have become a research focus in personalized medicine. We investigate the effects of biomarker misclassification on the design and analysis of stratified biomarker clinical trials. For a variety of inference problems including marker-treatment interaction in particular, we show that marker misclassification may have profound adverse effects on the coverage of confidence intervals, power of the tests, and required sample sizes. For each inferential problem, we propose methods to adjust for the classification errors.
Collapse
Affiliation(s)
- Chunling Liu
- Department of Applied Mathematics, Hong Kong Polytechnic University, Hong Kong, China
| | | | | | | | | |
Collapse
|
180
|
Identification of a genetic interaction between the tumor suppressor EAF2 and the retinoblastoma protein (Rb) signaling pathway in C. elegans and prostate cancer cells. Biochem Biophys Res Commun 2014; 447:292-8. [PMID: 24727455 DOI: 10.1016/j.bbrc.2014.03.138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 12/25/2022]
Abstract
The tumor suppressor EAF2 is regulated by androgen signaling and associated with prostate cancer. While EAF2 and its partner ELL have been shown to be members of protein complexes involved in RNA polymerase II transcriptional elongation, the biologic roles for EAF2 especially with regards to the development of cancer remains poorly understood. We have previously identified the eaf-1 gene in Caenorhabditiselegans as the ortholog of EAF2, and shown that eaf-1 interacts with the ELL ortholog ell-1 to control development and fertility in worms. To identify genetic pathways that interact with eaf-1, we screened RNAi libraries consisting of transcription factors, phosphatases, and chromatin-modifying factors to identify genes which enhance the effects of eaf-1(tm3976) on fertility. From this screen, we identified lin-53, hmg-1.2, pha-4, ruvb-2 and set-6 as hits. LIN-53 is the C. elegans ortholog of human retinoblastoma binding protein 4/7 (RBBP 4/7), which binds to the retinoblastoma protein and inhibits the Ras signaling pathway. We find that lin-53 showed a synthetic interaction with eaf-1(tm3976) where knockdown of lin-53 in an eaf-1(tm3976) mutant resulted in sterile worms. This phenotype may be due to cell death as the treated worms contain degenerated embryos with increased expression of the ced-1:GFP cell death marker. Further we find that the interaction between eaf-1 and lin-53/RBBP4/7 also exists in vertebrates, which is reflected by the formation of a protein complex between EAF2 and RBBP4/7. Finally, overexpression of either human EAF2 or RBBP4 in LNCaP cells induced the cell death while knockdown of EAF2 in LNCaP enhanced cell proliferation, indicating an important role of EAF2 in controlling the growth and survival of prostate cancer cells. Together these findings identify a novel physical and functional interaction between EAF2 and the Rb pathway.
Collapse
|
181
|
Beltran H, Tomlins S, Aparicio A, Arora V, Rickman D, Ayala G, Huang J, True L, Gleave ME, Soule H, Logothetis C, Rubin MA. Aggressive variants of castration-resistant prostate cancer. Clin Cancer Res 2014; 20:2846-50. [PMID: 24727321 DOI: 10.1158/1078-0432.ccr-13-3309] [Citation(s) in RCA: 326] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A subset of patients with advanced castration-resistant prostate cancer may eventually evolve into an androgen receptor (AR)-independent phenotype, with a clinical picture associated with the development of rapidly progressive disease involving visceral sites and hormone refractoriness, often in the setting of a low or modestly rising serum prostate-specific antigen level. Biopsies performed in such patients may vary, ranging from poorly differentiated carcinomas to mixed adenocarcinoma-small cell carcinomas to pure small cell carcinomas. These aggressive tumors often demonstrate low or absent AR protein expression and, in some cases, express markers of neuroendocrine differentiation. Because tumor morphology is not always predicted by clinical behavior, the terms "anaplastic prostate cancer" or "neuroendocrine prostate cancer" have been used descriptively to describe these rapidly growing clinical features. Patients meeting clinical criteria of anaplastic prostate cancer have been shown to predict for poor prognosis, and these patients may be considered for platinum-based chemotherapy treatment regimens. Therefore, understanding variants within the spectrum of advanced prostate cancer has important diagnostic and treatment implications.
Collapse
Affiliation(s)
- Himisha Beltran
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, CanadaAuthors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Scott Tomlins
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Ana Aparicio
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Vivek Arora
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - David Rickman
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, CanadaAuthors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Gustavo Ayala
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Jiaoti Huang
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Lawrence True
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Martin E Gleave
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Howard Soule
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Christopher Logothetis
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Mark A Rubin
- Authors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, CanadaAuthors' Affiliations: Division of Hematology and Medical Oncology; Institute for Precision Medicine, New York Presbyterian; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College; Department of Oncology, Memorial Sloan Kettering, New York, New York; Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Oncology, The University of Texas MD Anderson Cancer Center; Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center, Houston, Texas; Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles; Prostate Cancer Foundation, Santa Monica, California; Department of Pathology, University of Washington, Seattle, Washington; and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
182
|
Ragazzon B, Libé R, Assié G, Tissier F, Barreau O, Houdayer C, Perlemoine K, Audebourg A, Clauser E, René-Corail F, Bertagna X, Dousset B, Bertherat J, Groussin L. Mass-array screening of frequent mutations in cancers reveals RB1 alterations in aggressive adrenocortical carcinomas. Eur J Endocrinol 2014; 170:385-91. [PMID: 24347427 DOI: 10.1530/eje-13-0778] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CONTEXT Adrenocortical carcinoma (ACC) is a rare disease with a poor overall outcome. Transcriptome analysis identified two groups of ACCs with different prognosis. In aggressive ACCs, somatic mutations of the tumor suppressor gene TP53 and the proto-oncogene β-catenin are detected in 50% of cases. For the remaining aggressive ACCs and for the group with a better prognosis, molecular alterations are unknown. OBJECTIVE To identify new molecular actors driving adrenal tumorigenesis. EXPERIMENTAL DESIGN Analysis by mass array of 374 mutations among 32 common oncogenes or tumor suppressor genes was performed on the tumoral DNA of 26 ACCs, using Sequenom OncoCarta Panels. RESULTS Four mutations were identified, two previously known β-catenin mutations and one alteration in two other genes: JAK3 and retinoblastoma gene (RB1). The JAK3 alteration was found in leukocyte DNA and therefore considered as a polymorphism and not a somatic event. The full RB1 tumor suppressor gene was subsequently sequenced in a cohort of 49 ACCs (26 ACCs from the 'OncoCarta cohort' and 23 other ACCs): three somatic mutations were identified, all in the poor-outcome ACC group. By immunohistochemistry, a loss of the retinoblastoma protein (pRb) was found exclusively in aggressive ACCs in 27% of cases (seven out of 26), three of them with an inactivating RB1 mutation. Among the seven pRb-negative ACCs, five had an allele loss at the RB1 locus. CONCLUSIONS Parallel analysis of somatic mutations among known cancer genes allowed us to identify RB1 as a new actor in aggressive ACCs. These results suggest a prognostic significance of pRb expression loss in ACCs.
Collapse
|
183
|
Cereda V, Formica V, Massimiani G, Tosetto L, Roselli M. Targeting metastatic castration-resistant prostate cancer: mechanisms of progression and novel early therapeutic approaches. Expert Opin Investig Drugs 2014; 23:469-87. [PMID: 24490883 DOI: 10.1517/13543784.2014.885950] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Advances in clinical research have led to official approval of several new treatments for metastatic prostate cancer in the last three years: sipuleucel-T, cabazitaxel, abiraterone acetate, radium-223 and enzalutamide. Although these agents have all been shown to improve overall survival in randomized Phase III trials, metastatic castration-resistant prostate cancer (mCRPC) remains incurable. AREAS COVERED First, the review summarizes the current literature on the biology of mCRPC. The emerging data are increasing our understanding of the mechanisms that underlie the pathogenesis of castrate resistance and where future treatment might be headed. In the second part of the review, the authors assess the future directions in disease therapy. Indeed, novel selected therapeutic approaches, including novel agents and combinatorial therapies, are showing promising early results. EXPERT OPINION Targeting different molecular pathways in combination with immunotherapy can be a promising direction in metastatic castration prostate cancer treatment. However, several challenges still exist including elucidating the optimal use and sequencing of these new agents. There are also challenges in both the design and the interpretation of the results from clinical trials.
Collapse
Affiliation(s)
- Vittore Cereda
- University of Rome 'Tor Vergata', Tor Vergata Clinical Center, Department of Systems Medicine, Medical Oncology , V.le Oxford 81, 00133, Rome , Italy +390 620 908 190 ; +390 620 904 576 ;
| | | | | | | | | |
Collapse
|
184
|
Tan HL, Sood A, Rahimi HA, Wang W, Gupta N, Hicks J, Mosier S, Gocke CD, Epstein JI, Netto GJ, Liu W, Isaacs WB, De Marzo AM, Lotan TL. Rb loss is characteristic of prostatic small cell neuroendocrine carcinoma. Clin Cancer Res 2013; 20:890-903. [PMID: 24323898 DOI: 10.1158/1078-0432.ccr-13-1982] [Citation(s) in RCA: 257] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Small cell neuroendocrine carcinoma of the prostate is likely to become increasingly common with recent advances in pharmacologic androgen suppression. Thus, developing molecular markers of small cell differentiation in prostate cancer will be important to guide the diagnosis and therapy of this aggressive tumor. EXPERIMENTAL DESIGN We examined the status of RB1, TP53, and PTEN in prostatic small cell and acinar carcinomas via immunohistochemistry (IHC), copy-number alteration analysis, and sequencing of formalin-fixed paraffin-embedded specimens. RESULTS We found retinoblastoma (Rb) protein loss in 90% of small cell carcinoma cases (26 of 29) with RB1 allelic loss in 85% of cases (11 of 13). Of acinar tumors occurring concurrently with prostatic small cell carcinoma, 43% (3 of 7) showed Rb protein loss. In contrast, only 7% of primary high-grade acinar carcinomas (10 of 150), 11% of primary acinar carcinomas with neuroendocrine differentiation (4 of 35), and 15% of metastatic castrate-resistant acinar carcinomas (2 of 13) showed Rb protein loss. Loss of PTEN protein was seen in 63% of small cell carcinomas (17 of 27), with 38% (5 of 13) showing allelic loss. By IHC, accumulation of p53 was observed in 56% of small cell carcinomas (14 of 25), with 60% of cases (6 of 10) showing TP53 mutation. CONCLUSIONS Loss of RB1 by deletion is a common event in prostatic small cell carcinoma and can be detected by a validated IHC assay. As Rb protein loss rarely occurs in high-grade acinar tumors, these data suggest that Rb loss is a critical event in the development of small cell carcinomas and may be a useful diagnostic and potential therapeutic target.
Collapse
Affiliation(s)
- Hsueh-Li Tan
- Authors' Affiliations: Pathology, Department of Pathology, Henry Ford Health System, Detroit, Michigan; Oncology, and Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland; and Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Boland ML, Chourasia AH, Macleod KF. Mitochondrial dysfunction in cancer. Front Oncol 2013; 3:292. [PMID: 24350057 PMCID: PMC3844930 DOI: 10.3389/fonc.2013.00292] [Citation(s) in RCA: 333] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/17/2013] [Indexed: 12/18/2022] Open
Abstract
A mechanistic understanding of how mitochondrial dysfunction contributes to cell growth and tumorigenesis is emerging beyond Warburg as an area of research that is under-explored in terms of its significance for clinical management of cancer. Work discussed in this review focuses less on the Warburg effect and more on mitochondria and how dysfunctional mitochondria modulate cell cycle, gene expression, metabolism, cell viability, and other established aspects of cell growth and stress responses. There is increasing evidence that key oncogenes and tumor suppressors modulate mitochondrial dynamics through important signaling pathways and that mitochondrial mass and function vary between tumors and individuals but the significance of these events for cancer are not fully appreciated. We explore the interplay between key molecules involved in mitochondrial fission and fusion and in apoptosis, as well as in mitophagy, biogenesis, and spatial dynamics of mitochondria and consider how these distinct mechanisms are coordinated in response to physiological stresses such as hypoxia and nutrient deprivation. Importantly, we examine how deregulation of these processes in cancer has knock on effects for cell proliferation and growth. We define major forms of mitochondrial dysfunction and address the extent to which the functional consequences of such dysfunction can be determined and exploited for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Michelle L Boland
- The Ben May Department for Cancer Research, The University of Chicago , Chicago, IL , USA ; Committee on Molecular Metabolism and Nutrition, The University of Chicago , Chicago, IL , USA
| | - Aparajita H Chourasia
- The Ben May Department for Cancer Research, The University of Chicago , Chicago, IL , USA ; Committee on Cancer Biology, The University of Chicago , Chicago, IL , USA
| | - Kay F Macleod
- The Ben May Department for Cancer Research, The University of Chicago , Chicago, IL , USA ; Committee on Molecular Metabolism and Nutrition, The University of Chicago , Chicago, IL , USA ; Committee on Cancer Biology, The University of Chicago , Chicago, IL , USA
| |
Collapse
|
186
|
Song Y, Gilbert D, O’Sullivan TN, Yang C, Pan W, Fathalizadeh A, Lu L, Haines DC, Martin PL, Van Dyke T. Carcinoma initiation via RB tumor suppressor inactivation: a versatile approach to epithelial subtype-dependent cancer initiation in diverse tissues. PLoS One 2013; 8:e80459. [PMID: 24312475 PMCID: PMC3846618 DOI: 10.1371/journal.pone.0080459] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/03/2013] [Indexed: 01/02/2023] Open
Abstract
Carcinomas arise in a complex microenvironment consisting of multiple distinct epithelial lineages surrounded by a variety of stromal cell types. Understanding cancer etiologies requires evaluating the relationship among cell types during disease initiation and through progression. Genetically engineered mouse (GEM) models facilitate the prospective examination of early oncogenic events, which is not possible in humans. Since most solid tumors harbor aberrations in the RB network, we developed an inducible GEM approach for the establishment and assessment of carcinoma initiation in a diverse range of epithelial tissues and subtypes upon inactivation of RB-mediated tumor suppression (RB-TS). The system allows independent assessment of epithelial subtypes that express either cytokeratins (K) 18 or 19. By Cre-dependent expression of a protein that dominantly inactivates RB and functionally redundant proteins p107 and p130, neoplasia could be initiated in either K18 or K19 expressing cells of numerous tissues. By design, because only a single pathway aberration was engineered, carcinomas developed stochastically only after long latency. Hence, this system, which allows for directed cell type-specific carcinoma initiation, facilitates further definition of events that can progress neoplasms to aggressive cancers via engineered, carcinogen-induced and/or spontaneous evolution.
Collapse
Affiliation(s)
- Yurong Song
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Debra Gilbert
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - T. Norene O’Sullivan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Chunyu Yang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Wenqi Pan
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | - Lucy Lu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Diana C. Haines
- Pathology/Histotechnology Laboratory, SAIC-Frederick, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Philip L. Martin
- Center for Advanced Preclinical Research, SAIC-Frederick, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Terry Van Dyke
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| |
Collapse
|
187
|
Thelen P, Heinrich E, Bremmer F, Trojan L, Strauss A. Testosterone boosts for treatment of castration resistant prostate cancer: an experimental implementation of intermittent androgen deprivation. Prostate 2013; 73:1699-709. [PMID: 23868789 DOI: 10.1002/pros.22711] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 06/13/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND The primary therapeutic target for non-organ-confined prostate cancer is the androgen receptor (AR). Main strategies to ablate AR function are androgen depletion and direct receptor blockade by AR antagonists. However, incurable castration resistant prostate cancer (CRPC) develops resistance mechanisms to cope with trace amounts of androgen including AR overexpression and mutation in the AR ligand binding domain. METHODS The CRPC cell model VCaP derivative of a prostate cancer bone metastasis was used in vitro and in nude mice in vivo to examine the effects of immediate testosterone boost on CRPC cells. In addition, a testosterone tolerant cell model was established by incremental acclimatization of VCaP cells to 1 nM testosterone. The effects of androgen withdrawal and testosterone boosts on gene expression were assessed by quantitative real-time polymerase chain reaction, ELISA, and Western blots. Tumor cell proliferation was evaluated with a BrdU test. RESULTS Testosterone boosts on CRPC VCaP cells eliminate tumor cells to a higher extent than androgen withdrawal in androgen tolerant cells. The pronounced decrease of tumor cell proliferation was accompanied by a marked downregulation of AR expression regarding full-length AR and splice variant AR V7. CONCLUSIONS Acquiring castration resistance of prostate cancer cells by AR overexpression and amplification obviously sensitizes such cells to testosterone concentrations as low as physiological values. This introduces novel therapeutic means to treat CRPC with non-toxic measures and may find clinical implementation in intermittent androgen deprivation regimens.
Collapse
Affiliation(s)
- Paul Thelen
- Department of Urology, University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | |
Collapse
|
188
|
Goodwin JF, Schiewer MJ, Dean JL, Schrecengost RS, de Leeuw R, Han S, Ma T, Den RB, Dicker AP, Feng FY, Knudsen KE. A hormone-DNA repair circuit governs the response to genotoxic insult. Cancer Discov 2013; 3:1254-71. [PMID: 24027197 PMCID: PMC3823813 DOI: 10.1158/2159-8290.cd-13-0108] [Citation(s) in RCA: 282] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED Alterations in DNA repair promote tumor development, but the impact on tumor progression is poorly understood. Here, discovery of a biochemical circuit linking hormone signaling to DNA repair and therapeutic resistance is reported. Findings show that androgen receptor (AR) activity is induced by DNA damage and promotes expression and activation of a gene expression program governing DNA repair. Subsequent investigation revealed that activated AR promotes resolution of double-strand breaks and resistance to DNA damage both in vitro and in vivo. Mechanistically, DNA-dependent protein kinase catalytic subunit (DNAPKcs) was identified as a key target of AR after damage, controlling AR-mediated DNA repair and cell survival after genotoxic insult. Finally, DNAPKcs was shown to potentiate AR function, consistent with a dual role in both DNA repair and transcriptional regulation. Combined, these studies identify the AR-DNAPKcs circuit as a major effector of DNA repair and therapeutic resistance and establish a new node for therapeutic intervention in advanced disease. SIGNIFICANCE The present study identifies for the fi rst time a positive feedback circuit linking hormone action to the DNA damage response and shows the significant impact of this process on tumor progression and therapeutic response. These provocative findings provide the foundation for development of novel nodes of therapeutic intervention for advanced disease.
Collapse
Affiliation(s)
- Jonathan F. Goodwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Matthew J. Schiewer
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jeffry L. Dean
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Randy S. Schrecengost
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Renee de Leeuw
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sumin Han
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Teng Ma
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Robert B. Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam P. Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Felix Y. Feng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan*
| | - Karen E. Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
189
|
Liu C, Wang C, Wang K, Liu L, Shen Q, Yan K, Sun X, Chen J, Liu J, Ren H, Liu H, Xu Z, Hu S, Xu D, Fan Y. SMYD3 as an oncogenic driver in prostate cancer by stimulation of androgen receptor transcription. J Natl Cancer Inst 2013; 105:1719-28. [PMID: 24174655 DOI: 10.1093/jnci/djt304] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Androgen receptor (AR) is critical for prostate tumorigenesis and is frequently overexpressed during prostate cancer (PC) progression. However, few studies have addressed the epigenetic regulation of AR expression. METHODS We analyzed SMYD3 expression in human PC with Western blot and immunohistochemistry. SMYD3 expression was knocked down using short hairpin RNA (shRNA) or small interfering RNA (siRNA). Cell proliferation, colony formation, and apoptosis analyses and xenograft transplantation were performed to evaluate the impact of SMYD3 depletion on PC cells. AR expression and promoter activity were determined using real-time quantitative polymerase chain reaction, western blot, and luciferase reporter assay. AR promoter association with Sp1, SMYD3, and histone modifications was assessed by chromatin immunoprecipitation. Differences in AR mRNA abundance and promoter activity were analyzed using Wilcoxon signed-rank tests, SMYD3 expression was analyzed using with Mann-Whitney U tests for unpaired samples, and tumor weight was analyzed with Student t test. All statistical tests were two-sided. RESULTS The upregulation of SMYD3 protein expression was observed in seven of eight prostate tumor specimens, compared with matched normal tissues. Immunohistochemical analysis showed a strong SMYD3 staining in the nuclei of PC tissues in eight of 25 (32%) cases and in the cytoplasm in 23 out of 25 (92%) cases, whereas benign prostate tissue exhibited weak immunostaining. Depletion of SMYD3 by siRNA or shRNA inhibited PC cell proliferation (72 hours relative to 24 hours: control shRNA vs SMYD3 shRNA 1: mean fold change = 2.76 vs 1.68; difference = 1.08; 95% confidence interval = 0.78 to 1.38, P < .001), colony formation, cell migration, invasion, and xenograft tumor formation. Two functional SMYD3-binding motifs were identified in the AR promoter region. CONCLUSIONS SMYD3 promotes prostate tumorigenesis and mediates epigenetic upregulation of AR expression.
Collapse
Affiliation(s)
- Cheng Liu
- Affiliations of authors: Department of Urology (CL, CW, KW, KY, JC, JL, HR, HL, ZX, YF), Department of General Surgery (LL, SH), and School of Nursing (LL), Shandong University Qilu Hospital, Jinan, Shandong, China; Central Research Laboratory of Shandong University Second Hospital, Jinan, Shandong, China (DX); Department of Urology, Peking University First Hospital, Beijing, China (QS); Department of Urology, Qingdao Municipal Hospital, Qingdao, China (XS); Department of Medicine, Division of Haematology and Centre for Molecular Medicine, Karolinska University Hospital Solna and Karolinska Institutet, Stockholm, Sweden (CW, DX)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Qu X, Randhawa G, Friedman C, Kurland BF, Glaskova L, Coleman I, Mostaghel E, Higano CS, Porter C, Vessella R, Nelson PS, Fang M. A three-marker FISH panel detects more genetic aberrations of AR, PTEN and TMPRSS2/ERG in castration-resistant or metastatic prostate cancers than in primary prostate tumors. PLoS One 2013; 8:e74671. [PMID: 24098661 PMCID: PMC3787014 DOI: 10.1371/journal.pone.0074671] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/04/2013] [Indexed: 12/31/2022] Open
Abstract
TMPRSS2/ERG rearrangement, PTEN gene deletion, and androgen receptor (AR) gene amplification have been observed in various stages of human prostate cancer. We hypothesized that using these markers as a combined panel would allow better differentiation between low-risk and high-risk prostate cancer. We analyzed 110 primary prostate cancer samples, 70 metastatic tumor samples from 11 patients, and 27 xenograft tissues derived from 22 advanced prostate cancer patients using fluorescence in situ hybridization (FISH) analysis with probes targeting the TMPRSS2/ERG, PTEN, and AR gene loci. Heterogeneity of the aberrations detected was evaluated. Genetic patterns were also correlated with transcript levels. Among samples with complete data available, the three-marker FISH panel detected chromosomal abnormalities in 53% of primary prostate cancers and 87% of metastatic (Met) or castration-resistant (CRPC) tumors. The number of markers with abnormal FISH result had a different distribution between the two groups (P<0.001). At the patient level, Met/CRPC tumors are 4.5 times more likely to show abnormalities than primary cancer patients (P<0.05). Heterogeneity among Met/CRPC tumors is mostly inter-patient. Intra-patient heterogeneity is primarily due to differences between the primary prostate tumor and the metastases while multiple metastatic sites show consistent abnormalities. Intra-tumor variability is most prominent with the AR copy number in primary tumors. AR copy number correlated well with the AR mRNA expression (rho = 0.52, P<0.001). Especially among TMPRSS2:ERG fusion-positive CRPC tumors, AR mRNA and ERG mRNA levels are strongly correlated (rho = 0.64, P<0.001). Overall, the three-marker FISH panel may represent a useful tool for risk stratification of prostate cancer patients.
Collapse
Affiliation(s)
- Xiaoyu Qu
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Grace Randhawa
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
| | - Cynthia Friedman
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
| | - Brenda F. Kurland
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lena Glaskova
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
| | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Elahe Mostaghel
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - Celestia S. Higano
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - Christopher Porter
- Virginia Mason Medical Center, Seattle, Washington, United States of America
| | - Robert Vessella
- University of Washington, Seattle, Washington, United States of America
- Puget Sound VA Health Care System, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - Min Fang
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
191
|
Egan A, Dong Y, Zhang H, Qi Y, Balk SP, Sartor O. Castration-resistant prostate cancer: adaptive responses in the androgen axis. Cancer Treat Rev 2013; 40:426-33. [PMID: 24139549 DOI: 10.1016/j.ctrv.2013.09.011] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/06/2013] [Indexed: 12/22/2022]
Abstract
The androgen signaling axis in prostate cancer is associated with multiple adaptive mechanisms in response to castration. Herein we review these adaptations with an emphasis on recent molecular insights into the growth and development of castration resistant prostate cancer (CRPC). Alterations include both conventional and novel intracrine androgen synthesis pathways and androgen transport as well as androgen receptor (AR) overexpression, mutation, and splice variation. Each of these underlying mechanisms are potentially linked to post-castration growth, especially after treatment with newer hormonal agents such as abiraterone and enzalutamide. Post-translational AR modifications are well documented and these can affect receptor activity, stability, localization, and interaction with other proteins. Changes in recruitment of androgen receptor associated co-activators/repressors and a distinct AR-induced transcriptional program can dramatically alter proliferation, invasion, and metastasis in a ligand and context-dependent manner. Numerous previously uncharacterized non-coding RNAs, some of which are androgen regulated, may also have important biological function in this disease. Taken together, the view of CRPC has changed dramatically in the last several years. This has occurred not only within the setting of multiple treatment paradigm changes, but also as a multiplicity of potential molecular mechanisms underlying this disease state have been explored and discovered.
Collapse
Affiliation(s)
- Alison Egan
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yan Dong
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; National Engineering Laboratory for AIDS Vaccine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Haitao Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yanfeng Qi
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Steven P Balk
- College of Life Sciences, Jilin University, China and Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Urology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Department of Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
192
|
Engelmann D, Mayoli-Nüssle D, Mayrhofer C, Fürst K, Alla V, Stoll A, Spitschak A, Abshagen K, Vollmar B, Ran S, Pützer BM. E2F1 promotes angiogenesis through the VEGF-C/VEGFR-3 axis in a feedback loop for cooperative induction of PDGF-B. J Mol Cell Biol 2013; 5:391-403. [PMID: 24014887 DOI: 10.1093/jmcb/mjt035] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is essential for primary tumor growth and metastatic dissemination. E2F1, frequently upregulated in advanced cancers, was recently shown to drive malignant progression. In an attempt to decipher the molecular events underlying this behavior, we demonstrate that the tumor cell-associated vascular endothelial growth factor-C/receptor-3 (VEGF-C/VEGFR-3) axis is controlled by E2F1. Activation or forced expression of E2F1 in cancer cells leads to the upregulation of VEGFR-3 and its ligand VEGF-C, whereas E2F1 depletion prevents their expression. E2F1-dependent receptor induction is crucial for tumor cells to enhance formation of capillary tubes and neovascularization in mice. We further provide evidence for a positive feedback loop between E2F1 and VEGFR-3 signaling to stimulate pro-angiogenic platelet-derived growth factor B (PDGF-B). E2F1 or VEGFR-3 knockdown results in reduced PDGF-B levels, while the coexpression synergistically upregulates promoter activity and endogenous protein expression of PDGF-B. Our findings delineate an as yet unrecognized function of E2F1 as enhancer of angiogenesis via regulation of VEGF-C/VEGFR-3 signaling in tumors to cooperatively activate PDGF-B expression. Targeting this pathway might be reasonable to complement standard anti-angiogenic treatment of cancers with deregulated E2F1.
Collapse
Affiliation(s)
- David Engelmann
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Abstract
Prostate cancer (PCa) is the most commonly diagnosed noncutaneous malignancy and second leading cause of cancer-related deaths in US males. Clinically, locally confined disease is treated surgically and/or with radiation therapy. Invasive disease, however, must be treated with pharmacological inhibitors of androgen receptor (AR) activity, since disease progression is fundamentally reliant on AR activation. However, despite initially effective treatment options, recurrent castration-resistant PCa (CRPC) often occurs due to aberrant reactivation of AR. Additionally, it is appreciated that many other signaling molecules, such as transcription factors, oncogenes, and tumor suppressors, are often perturbed and significantly contribute to PCa initiation and progression to incurable disease. Understanding the interplay between AR signaling and other signaling networks altered in PCa will advance therapeutic approaches. Overall, comprehension of the molecular composition promoting neoplastic growth and formation of CRPC is paramount for developing durable treatment options.
Collapse
Affiliation(s)
- Randy Schrecengost
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | |
Collapse
|
194
|
Su S, Minges JT, Grossman G, Blackwelder AJ, Mohler JL, Wilson EM. Proto-oncogene activity of melanoma antigen-A11 (MAGE-A11) regulates retinoblastoma-related p107 and E2F1 proteins. J Biol Chem 2013; 288:24809-24. [PMID: 23853093 DOI: 10.1074/jbc.m113.468579] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Melanoma antigen-A11 (MAGE-A11) is a low-abundance, primate-specific steroid receptor coregulator in normal tissues of the human reproductive tract that is expressed at higher levels in prostate cancer. Increased expression of MAGE-A11 enhances androgen receptor transcriptional activity and promotes prostate cancer cell growth. Further investigation into the mechanisms of MAGE-A11 function in prostate cancer demonstrated interactions with the retinoblastoma-related protein p107 and Rb tumor suppressor but no interaction with p130 of the Rb family. MAGE-A11 interaction with p107 was associated with transcriptional repression in cells with low MAGE-A11 and transcriptional activation in cells with higher MAGE-A11. Selective interaction of MAGE-A11 with retinoblastoma family members suggested the regulation of E2F transcription factors. MAGE-A11 stabilized p107 by inhibition of ubiquitination and linked p107 to hypophosphorylated E2F1 in association with the stabilization and activation of E2F1. The androgen receptor and MAGE-A11 modulated endogenous expression of the E2F1-regulated cyclin-dependent kinase inhibitor p27(Kip1). The ability of MAGE-A11 to increase E2F1 transcriptional activity was similar to the activity of adenovirus early oncoprotein E1A and depended on MAGE-A11 interactions with p107 and p300. The immunoreactivity of p107 and MAGE-A11 was greater in advanced prostate cancer than in benign prostate, and knockdown with small inhibitory RNA showed that p107 is a transcriptional activator in prostate cancer cells. These results suggest that MAGE-A11 is a proto-oncogene whose increased expression in prostate cancer reverses retinoblastoma-related protein p107 from a transcriptional repressor to a transcriptional activator of the androgen receptor and E2F1.
Collapse
Affiliation(s)
- Shifeng Su
- Laboratories for Reproductive Biology, Department of Pediatrics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | |
Collapse
|
195
|
Burton DGA, Giribaldi MG, Munoz A, Halvorsen K, Patel A, Jorda M, Perez-Stable C, Rai P. Androgen deprivation-induced senescence promotes outgrowth of androgen-refractory prostate cancer cells. PLoS One 2013; 8:e68003. [PMID: 23840802 PMCID: PMC3695935 DOI: 10.1371/journal.pone.0068003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 05/28/2013] [Indexed: 01/07/2023] Open
Abstract
Androgen deprivation (AD) is an effective method for initially suppressing prostate cancer (PC) progression. However, androgen-refractory PC cells inevitably emerge from the androgen-responsive tumor, leading to incurable disease. Recent studies have shown AD induces cellular senescence, a phenomenon that is cell-autonomously tumor-suppressive but which confers tumor-promoting adaptations that can facilitate the advent of senescence-resistant malignant cell populations. Because androgen-refractory PC cells emerge clonally from the originally androgen-responsive tumor, we sought to investigate whether AD-induced senescence (ADIS) affects acquisition of androgen-refractory behavior in androgen-responsive LNCaP and LAPC4 prostate cancer cells. We find that repeated exposure of these androgen-responsive cells to senescence-inducing stimuli via cyclic AD leads to the rapid emergence of ADIS-resistant, androgen-refractory cells from the bulk senescent cell population. Our results show that the ADIS phenotype is associated with tumor-promoting traits, notably chemoresistance and enhanced pro-survival mechanisms such as inhibition of p53-mediated cell death, which encourage persistence of the senescent cells. We further find that pharmacologic enforcement of p53/Bax activation via Nutlin-3 prior to establishment of ADIS is required to overcome the associated pro-survival response and preferentially trigger pervasive cell death instead of senescence during AD. Thus our study demonstrates that ADIS promotes outgrowth of androgen-refractory PC cells and is consequently a suboptimal tumor-suppressor response to AD.
Collapse
Affiliation(s)
- Dominick G. A. Burton
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Maria G. Giribaldi
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Biology Department, University of Miami, Coral Gables, Florida, United States of America
| | - Anisleidys Munoz
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Biology Department, University of Miami, Coral Gables, Florida, United States of America
| | - Katherine Halvorsen
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Asmita Patel
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Merce Jorda
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Carlos Perez-Stable
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Bruce W. Carter Veterans Affairs Medical Center, Miami, Florida, United States of America
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Florida, United States of America
| | - Priyamvada Rai
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Florida, United States of America
- * E-mail:
| |
Collapse
|
196
|
Prostate cancer progression after androgen deprivation therapy: mechanisms of castrate resistance and novel therapeutic approaches. Oncogene 2013; 32:5501-11. [PMID: 23752182 DOI: 10.1038/onc.2013.206] [Citation(s) in RCA: 586] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/10/2013] [Accepted: 04/10/2013] [Indexed: 12/13/2022]
Abstract
Prostate cancer is the second-leading cause of cancer-related mortality in men in Western societies. Androgen receptor (AR) signaling is a critical survival pathway for prostate cancer cells, and androgen-deprivation therapy (ADT) remains the principal treatment for patients with locally advanced and metastatic disease. Although a majority of patients initially respond to ADT, most will eventually develop castrate resistance, defined as disease progression despite serum testosterone levels of <20 ng/dl. The recent discovery that AR signaling persists during systemic castration via intratumoral production of androgens led to the development of novel anti-androgen therapies including abiraterone acetate and enzalutamide. Although these agents effectively palliate symptoms and prolong life, metastatic castration-resistant prostate cancer remains incurable. An increased understanding of the mechanisms that underlie the pathogenesis of castrate resistance is therefore needed to develop novel therapeutic approaches for this disease. The aim of this review is to summarize the current literature on the biology and treatment of castrate-resistant prostate cancer.
Collapse
|
197
|
Xie X, Kerrigan JE, Minko T, Garbuzenko O, Lee KC, Scarborough A, Abali EE, Budak-Alpdogan T, Johnson-Farley N, Banerjee D, Scotto KW, Bertino JR. Antitumor and modeling studies of a penetratin-peptide that targets E2F-1 in small cell lung cancer. Cancer Biol Ther 2013; 14:742-51. [PMID: 23792570 DOI: 10.4161/cbt.25184] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
E2F-1, a key transcription factor necessary for cell growth, DNA repair, and differentiation, is an attractive target for development of anticancer drugs in tumors that are E2F "oncogene addicted". We identified a peptide isolated from phage clones that bound tightly to the E2F-1 promoter consensus sequence. The peptide was coupled to penetratin to enhance cellular uptake. Modeling of the penetratin-peptide (PEP) binding to the DNA E2F-1 promoter demonstrated favorable interactions that also involved the participation of most of the penetratin sequence. The penetratin-peptide (PEP) demonstrated potent in vitro cytotoxic effects against a range of cancer cell lines, particularly against Burkitt lymphoma cells and small cell lung cancer (SCLC) cells. Further studies in the H-69 SCLC cell line showed that the PEP inhibited transcription of E2F-1 and also several important E2F-regulated enzymes involved in DNA synthesis, namely, thymidylate synthase, thymidine kinase, and ribonucleotide reductase. As the PEP was found to be relatively unstable in serum, it was encapsulated in PEGylated liposomes for in vivo studies. Treatment of mice bearing the human small cell lung carcinoma H-69 with the PEP encapsulated in PEGylated liposomes (PL-PEP) caused tumor regression without significant toxicity. The liposome encapsulated PEP has promise as an antitumor agent, alone or in combination with inhibitors of DNA synthesis.
Collapse
Affiliation(s)
- Xiaoqi Xie
- Department of Pharmacology and Medicine, Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Inoue K, Fry EA, Taneja P. Recent progress in mouse models for tumor suppressor genes and its implications in human cancer. Clin Med Insights Oncol 2013; 7:103-22. [PMID: 23843721 PMCID: PMC3682694 DOI: 10.4137/cmo.s10358] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Gain-of-function mutations in oncogenes and loss-of-function mutations in tumor suppressor genes (TSG) lead to cancer. In most human cancers, these mutations occur in somatic tissues. However, hereditary forms of cancer exist for which individuals are heterozygous for a germline mutation in a TSG locus at birth. The second allele is frequently inactivated by gene deletion, point mutation, or promoter methylation in classical TSGs that meet Knudson's two-hit hypothesis. Conversely, the second allele remains as wild-type, even in tumors in which the gene is haplo-insufficient for tumor suppression. This article highlights the importance of PTEN, APC, and other tumor suppressors for counteracting aberrant PI3K, β-catenin, and other oncogenic signaling pathways. We discuss the use of gene-engineered mouse models (GEMM) of human cancer focusing on Pten and Apc knockout mice that recapitulate key genetic events involved in initiation and progression of human neoplasia. Finally, the therapeutic potential of targeting these tumor suppressor and oncogene signaling networks is discussed.
Collapse
Affiliation(s)
- Kazushi Inoue
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Elizabeth A. Fry
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Pankaj Taneja
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Department of Cancer Biology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|
199
|
Abstract
The combination of expression patterns of AGR2 (anterior gradient 2) and CD10 by prostate cancer provided four phenotypes that correlated with clinical outcome. Based on immunophenotyping, CD10(low)AGR2(high), CD10(high)AGR2(high), CD10(low)AGR2(low), and CD10(high)AGR2(low) were distinguished. AGR2(+) tumors were associated with longer recurrence-free survival and CD10(+) tumors with shorter recurrence-free survival. In high-stage cases, the CD10(low)AGR2(high) phenotype was associated with a ninefold higher recurrence-free survival than the CD10(high)AGR2(low) phenotype. The CD10(high)AGR2(high) and CD10(low)AGR2(low) phenotypes were intermediate. The CD10(high)AGR2(low) phenotype was most frequent in high-grade primary tumors. Conversely, bone and other soft tissue metastases, and derivative xenografts, expressed more AGR2 and less CD10. AGR2 protein was readily detected in tumor metastases. The CD10(high)AGR2(low) phenotype in primary tumors is predictive of poor outcome; however, the CD10(low)AGR2(high) phenotype is more common in metastases. It appears that AGR2 has a protective function in primary tumors but may have a role in the distal spread of tumor cells.
Collapse
|
200
|
Targeting cell cycle and hormone receptor pathways in cancer. Oncogene 2013; 32:5481-91. [PMID: 23708653 PMCID: PMC3898261 DOI: 10.1038/onc.2013.83] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 01/29/2013] [Accepted: 02/01/2013] [Indexed: 12/21/2022]
Abstract
The cyclin/cyclin-dependent kinase (CDK)/retinoblastoma (RB)-axis is a critical modulator of cell cycle entry and is aberrant in many human cancers. New nodes of therapeutic intervention are needed that can delay or combat the onset of malignancies. The antitumor properties and mechanistic functions of PD-0332991 (PD; a potent and selective CDK4/6 inhibitor) were investigated using human prostate cancer (PCa) models and primary tumors. PD significantly impaired the capacity of PCa cells to proliferate by promoting a robust G1-arrest. Accordingly, key regulators of the G1-S cell cycle transition were modulated including G1 cyclins D, E and A. Subsequent investigation demonstrated the ability of PD to function in the presence of existing hormone-based regimens and to cooperate with ionizing radiation to further suppress cellular growth. Importantly, it was determined that PD is a critical mediator of PD action. The anti-proliferative impact of CDK4/6 inhibition was revealed through reduced proliferation and delayed growth using PCa cell xenografts. Finally, first-in-field effects of PD on proliferation were observed in primary human prostatectomy tumor tissue explants. This study shows that selective CDK4/6 inhibition, using PD either as a single-agent or in combination, hinders key proliferative pathways necessary for disease progression and that RB status is a critical prognostic determinant for therapeutic efficacy. Combined, these pre-clinical findings identify selective targeting of CDK4/6 as a bona fide therapeutic target in both early stage and advanced PCa and underscore the benefit of personalized medicine to enhance treatment response.
Collapse
|