651
|
Characterization of the genomic architecture and mutational spectrum of a small cell prostate carcinoma. Genes (Basel) 2014; 5:366-84. [PMID: 24823478 PMCID: PMC4094938 DOI: 10.3390/genes5020366] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/01/2014] [Accepted: 04/15/2014] [Indexed: 12/24/2022] Open
Abstract
We present the use of a series of laboratory, analytical and interpretation methods to investigate personalized cancer care for a case of small cell prostate carcinoma (SCPC), a rare and aggressive tumor with poor prognosis, for which the underlying genomic architecture and mutational spectrum has not been well characterized. We performed both SNP genotyping and exome sequencing of a Virchow node metastasis from a patient with SCPC. A variety of methods were used to analyze and interpret the tumor genome for copy number variation, loss of heterozygosity (LOH), somatic mosaicism and mutations in genes from known cancer pathways. The combination of genotyping and exome sequencing approaches provided more information than either technique alone. The results showed widespread evidence of copy number changes involving most chromosomes including the possible loss of both alleles of CDKN1B (p27/Kip1). LOH was observed for the regions encompassing the tumor suppressors TP53, RB1, and CHD1. Predicted damaging somatic mutations were observed in the retained TP53 and RB1 alleles. Mutations in other genes that may be functionally relevant were noted, especially the recently reported high confidence cancer drivers FOXA1 and CCAR1. The disruption of multiple cancer drivers underscores why SCPC may be such a difficult cancer to manage.
Collapse
|
652
|
Characterization of prostate neuroendocrine cancers and therapeutic management: a literature review. Prostate Cancer Prostatic Dis 2014; 17:220-6. [DOI: 10.1038/pcan.2014.17] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/25/2014] [Accepted: 03/18/2014] [Indexed: 12/31/2022]
|
653
|
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
|
654
|
Krauss DJ, Amin M, Stone B, Ye H, Hayek S, Cotant M, Hafron J, Brabbins DS. Chromogranin A staining as a prognostic variable in newly diagnosed Gleason score 7-10 prostate cancer treated with definitive radiotherapy. Prostate 2014; 74:520-7. [PMID: 24375481 DOI: 10.1002/pros.22771] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/11/2013] [Indexed: 11/12/2022]
Abstract
PURPOSE To demonstrate the association of neuroendocrine differentiation, as identified by chromogranin A (CgA) staining, with clinical outcomes in newly diagnosed prostatic adenocarcinoma treated with definitive radiotherapy (RT). MATERIALS/METHODS Patients with Gleason score ≥7 adenocarcinoma were identified from our outcomes database. RT consisted of external beam, brachytherapy, or external beam with brachytherapy boost. Biopsy specimens were stained for neuroendocrine differentiation with CgA. Results were interpreted by a single pathologist. CgA staining was quantified as 0%, <1%, 1-10%, or >10% of tumor cells. Clinical outcomes were blinded at the time of pathologic evaluation. RESULTS CgA staining was performed on 289 patients. 149 patients had Gleason score 7, and 140 were Gleason score 8-10. Median follow-up was 6.5 years. For patients with <1% versus >1% CgA staining, pretreatment characteristics were well-balanced. CgA staining was detected in 90 cases (31%). 58 patients had focal positive (<1%) CgA staining, and 32 cases had >1% of tumor cells CgA positive. Patients with >1% CgA staining had inferior biochemical control, clinical failure, distant metastases (DM), and cause-specific survival (CSS) rates. Ten-year rates of DM were 8% versus 48% for patients with <1% versus >1% CgA positive cells, respectively (P < 0.001). CSS at 10 years was 95% versus 76%, respectively (P < 0.001). Local control was equivalent in the two patient cohorts. Patients with <1% CgA staining had similar outcomes to those patients with 0% staining. CONCLUSIONS Neuroendocrine differentiation involving >1% of tumor cells on prostate cancer biopsies is a predictor of DM and CSS in patients treated with primary RT.
Collapse
Affiliation(s)
- Daniel J Krauss
- Department of Radiation Oncology, Oakland University William Beaumont School of Medicine, Royal Oak, Michigan
| | | | | | | | | | | | | | | |
Collapse
|
655
|
Increased androgen receptor gene copy number is associated with TMPRSS2-ERG
rearrangement in prostatic small cell carcinoma. Mol Carcinog 2014; 54:900-7. [DOI: 10.1002/mc.22162] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/24/2014] [Accepted: 03/27/2014] [Indexed: 11/07/2022]
|
656
|
Fraser M, Berlin A, Bristow RG, van der Kwast T. Genomic, pathological, and clinical heterogeneity as drivers of personalized medicine in prostate cancer. Urol Oncol 2014; 33:85-94. [PMID: 24768356 DOI: 10.1016/j.urolonc.2013.10.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 12/23/2022]
Abstract
Prostate cancer (CaP) is the most commonly diagnosed malignancy in men in the Western world. In North America, more than 275,000 men are diagnosed annually, whereby approximately 1 in 6 men will be diagnosed with CaP in their lifetime, and 1 in 34 men will die from castration-resistant metastatic disease. Unfortunately, current clinical prognostic factors explain only a proportion of the observed variation in clinical outcome from patient to patient. Furthermore, overtreatment of indolent and low-risk cancers leads to inappropriate morbidity following radiotherapy or surgery. As such, better predictors of individualized prognosis and treatment response are urgently needed to triage patients to customized and intensified CaP treatment. Recent developments in next-generation sequencing have made it possible to identify prognostic and predictive signatures based on genomic profiles. We discuss the genetic basis of CaP progression from localized to systemic disease (e.g., point mutations, copy-number alterations, and structural variants) in relation with unique features of CaP biology, including intraprostatic and interprostatic heterogeneity, multifocality and multiclonality, TMPRSS2:ERG, and other ETS-family gene fusions. Finally, we focus on the use of genomic markers as prognostic factors for local failure and for systemic disease, as novel risk-stratification tools, in triaging patients to existing treatment options, and ultimately the potential of genomics for the identification of molecular targets for therapy of CaP.
Collapse
Affiliation(s)
- Michael Fraser
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada
| | - Alejandro Berlin
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Robert G Bristow
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Theodorus van der Kwast
- Department of Pathology and Laboratory Medicine, Toronto General Hospital (University Health Network), Toronto, Ontario, Canada.
| |
Collapse
|
657
|
Menon R, Deng M, Rüenauver K, Queisser A, Peifer M, Pfeifer M, Offermann A, Boehm D, Vogel W, Scheble V, Fend F, Kristiansen G, Wernert N, Oberbeckmann N, Biskup S, Rubin MA, Adler D, Perner S. Somatic copy number alterations by whole-exome sequencing implicates YWHAZ and PTK2 in castration-resistant prostate cancer. J Pathol 2014; 231:505-16. [PMID: 24114522 DOI: 10.1002/path.4274] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 09/10/2013] [Accepted: 09/19/2013] [Indexed: 01/03/2023]
Abstract
Castration-resistant prostate cancer (CRPC) is the most aggressive form of prostate cancer (PCa) and remains a significant therapeutic challenge. The key to the development of novel therapeutic targets for CRPC is to decipher the molecular alterations underlying this lethal disease. The aim of our study was to identify therapeutic targets for CRPC by assessing somatic copy number alterations (SCNAs) by whole-exome sequencing on five CRPC/normal paired formalin-fixed paraffin-embedded (FFPE) samples, using the SOLiD4 next-generation sequencing (NGS) platform. Data were validated using fluorescence in situ hybridization (FISH) on a PCa progression cohort. PTK2 and YWHAZ amplification, mRNA and protein expression were determined in selected PCa cell lines. Effects of PTK2 inhibition using TAE226 inhibitor and YWHAZ knock-down on cell proliferation and migration were tested in PC3 cells in vitro. In a larger validation cohort, the amplification frequency of YWHAZ was 3% in localized PCa and 48% in CRPC, whereas PTK2 was amplified in 1% of localized PCa and 35% in CRPC. YWHAZ knock-down and PTK2 inhibition significantly affected cell proliferation and migration in the PC3 cells. Our findings suggest that inhibition of YWHAZ and PTK2 could delay the progression of the disease in CRPC patients harbouring amplification of the latter genes. Furthermore, our validated whole-exome sequencing data show that FFPE tissue could be a promising alternative for SCNA screening using next-generation sequencing technologies.
Collapse
Affiliation(s)
- Roopika Menon
- Department of Prostate Cancer Research, University Hospital of Bonn, Germany; Institute of Pathology, University Hospital of Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
658
|
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: 321] [Impact Index Per Article: 32.1] [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
|
659
|
Terry S, Beltran H. The many faces of neuroendocrine differentiation in prostate cancer progression. Front Oncol 2014; 4:60. [PMID: 24724054 PMCID: PMC3971158 DOI: 10.3389/fonc.2014.00060] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 03/12/2014] [Indexed: 12/15/2022] Open
Abstract
In normal prostate, neuroendocrine (NE) cells are rare and interspersed among the epithelium. These cells are believed to provide trophic signals to epithelial cell populations through the secretion of an abundance of neuropeptides that can diffuse to influence surrounding cells. In the setting of prostate cancer (PC), NE cells can also stimulate surrounding prostate adenocarcinoma cell growth, but in some cases adenocarcinoma cells themselves acquire NE characteristics. This epithelial plasticity is associated with decreased androgen receptor (AR) signaling and the accumulation of neuronal and stem cell characteristics. Transformation to an NE phenotype is one proposed mechanism of resistance to contemporary AR-targeted treatments, is associated with poor prognosis, and thought to represent up to 25% of lethal PCs. Importantly, the advent of high-throughput technologies has started to provide clues for understanding the complex molecular profiles of tumors exhibiting NE differentiation. Here, we discuss these recent advances, the multifaceted manner by which an NE-like state may arise during the different stages of disease progression, and the potential benefit of this knowledge for the management of patients with advanced PC.
Collapse
Affiliation(s)
- Stéphane Terry
- U955, Institut Mondor de Recherche Biomédicale, INSERM , Créteil , France ; UMR 3244, Institut Curie , Paris , France
| | - Himisha Beltran
- Division of Hematology and Medical Oncology, Weill Cornell Medical College , New York, NY , USA
| |
Collapse
|
660
|
Beltran H. The N-myc Oncogene: Maximizing its Targets, Regulation, and Therapeutic Potential. Mol Cancer Res 2014; 12:815-22. [PMID: 24589438 DOI: 10.1158/1541-7786.mcr-13-0536] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
N-myc (MYCN), a member of the Myc family of basic-helix-loop-helix-zipper (bHLHZ) transcription factors, is a central regulator of many vital cellular processes. As such, N-myc is well recognized for its classic oncogenic activity and association with human neuroblastoma. Amplification and overexpression of N-myc has been described in other tumor types, particularly those of neural origin and neuroendocrine tumors. This review outlines N-myc's contribution to normal development and oncogenic progression. In addition, it highlights relevant transcriptional targets and mechanisms of regulation. Finally, the clinical implications of N-Myc as a biomarker and potential as a target using novel therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Himisha Beltran
- Author's Affiliation: Weill Cornell Medical College, New York, New York
| |
Collapse
|
661
|
Abstract
Pure small-cell carcinoma (SCC) of the prostate is a rare entity and one of the most aggressive malignancies of the prostate. Histologically, prostatic SCCs of the prostate are part of a spectrum of anaplastic tumours of the prostate and are similar to SCCs of the lungs. In most cases, SCC of the prostate is associated with conventional prostatic adenocarcinoma. Both components of these mixed tumours frequently share molecular alterations such as ERG gene rearrangements or AURKA and MYCN amplifications, suggesting a common clonal origin. The clinical behaviour of small-cell prostate carcinomas is characterized by extensive local disease, visceral disease, and low PSA levels despite large metastatic burden. Commonly, the emergence of the SCC occurs in patients with high-grade adenocarcinoma who are often treated with androgen deprivation treatment (ADT). However, SCCs do not usually benefit from ADT. A biopsy of accessible lesions is strongly recommended to identify those with SCC pathological features, as management is undoubtedly affected by this finding. Chemotherapy is the standard approach for treating patients with either localized or advanced prostatic SCC. Despite the emergence of more-aggressive treatment modalities, the prognosis of men with prostatic SCC remains dismal.
Collapse
|
662
|
Bristow RG, Berlin A, Dal Pra A. An arranged marriage for precision medicine: hypoxia and genomic assays in localized prostate cancer radiotherapy. Br J Radiol 2014; 87:20130753. [PMID: 24588670 DOI: 10.1259/bjr.20130753] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed malignancy in males in the Western world with one in six males diagnosed in their lifetime. Current clinical prognostication groupings use pathologic Gleason score, pre-treatment prostatic-specific antigen and Union for International Cancer Control-TNM staging to place patients with localized CaP into low-, intermediate- and high-risk categories. These categories represent an increasing risk of biochemical failure and CaP-specific mortality rates, they also reflect the need for increasing treatment intensity and justification for increased side effects. In this article, we point out that 30-50% of patients will still fail image-guided radiotherapy or surgery despite the judicious use of clinical risk categories owing to interpatient heterogeneity in treatment response. To improve treatment individualization, better predictors of prognosis and radiotherapy treatment response are needed to triage patients to bespoke and intensified CaP treatment protocols. These should include the use of pre-treatment genomic tests based on DNA or RNA indices and/or assays that reflect cancer metabolism, such as hypoxia assays, to define patient-specific CaP progression and aggression. More importantly, it is argued that these novel prognostic assays could be even more useful if combined together to drive forward precision cancer medicine for localized CaP.
Collapse
Affiliation(s)
- R G Bristow
- Princess Margaret Cancer Center (University Health Network), Toronto, ON, Canada
| | | | | |
Collapse
|
663
|
Cross modulation between the androgen receptor axis and protocadherin-PC in mediating neuroendocrine transdifferentiation and therapeutic resistance of prostate cancer. Neoplasia 2014; 15:761-72. [PMID: 23814488 DOI: 10.1593/neo.122070] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 04/14/2013] [Accepted: 04/15/2013] [Indexed: 12/31/2022] Open
Abstract
Castration-resistant prostate cancers (CRPCs) that relapse after androgen deprivation therapies (ADTs) are responsible for the majority of mortalities from prostate cancer (PCa). While mechanisms enabling recurrent activity of androgen receptor (AR) are certainly involved in the development of CRPC, there may be factors that contribute to the process including acquired neuroendocrine (NE) cell-like behaviors working through alternate (non-AR) cell signaling systems or AR-dependent mechanisms. In this study, we explore the potential relationship between the AR axis and a novel putative marker of NE differentiation, the human male protocadherin-PC (PCDH-PC), in vitro and in human situations. We found evidence for an NE transdifferentiation process and PCDH-PC expression as an early-onset adaptive mechanism following ADT and elucidate AR as a key regulator of PCDH-PC expression. PCDH-PC overexpression, in turn, attenuates the ligand-dependent activity of the AR, enabling certain prostate tumor clones to assume a more NE phenotype and promoting their survival under diverse stress conditions. Acquisition of an NE phenotype by PCa cells positively correlated with resistance to cytotoxic agents including docetaxel, a taxane chemotherapy approved for the treatment of patients with metastatic CRPC. Furthermore, knockdown of PCDH-PC in cells that have undergone an NE transdifferentiation partially sensitized cells to docetaxel. Together, these results reveal a reciprocal regulation between the AR axis and PCDH-PC signals, observed both in vitro and in vivo, with potential implications in coordinating NE transdifferentiation processes and progression of PCa toward hormonal and chemoresistance.
Collapse
|
664
|
Lorente D, De Bono JS. Molecular alterations and emerging targets in castration resistant prostate cancer. Eur J Cancer 2014; 50:753-64. [PMID: 24418724 DOI: 10.1016/j.ejca.2013.12.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 11/25/2013] [Accepted: 12/03/2013] [Indexed: 02/06/2023]
Abstract
Prostate cancer is the most common malignancy in Western Europe, of which approximately 10-20% presents with advanced or metastatic disease. Initial response with androgen deprivation therapy is almost universal, but progression to castration resistant prostate cancer (CRPC), an incurable disease, occurs in approximately 2-3 years. In recent years, the novel taxane cabazitaxel, the hormonal agents abiraterone and enzalutamide, the immunotherapeutic agent sipuleucel-T and the radiopharmaceutical radium-223 have been shown to prolong survival in large randomised trials, thus widely increasing the therapeutic armamentarium against the disease. Despite these advances, the median survival in the first-line setting of metastatic castration-resistant prostate cancer (mCRPC) is still up to 25 months and in the post-docetaxel setting is about 15-18 months. There is an urgent need for the development of biomarkers of treatment response, and for a deeper understanding of tumour heterogeneity and the molecular biology underlying the disease. In this review, we attempt to provide insight into the novel molecular targets showing promise in clinical trials.
Collapse
Affiliation(s)
- D Lorente
- Prostate Cancer Targeted Therapy Group and Drug Development Unit, The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, SM2 5PT Sutton, Surrey, UK
| | - J S De Bono
- Prostate Cancer Targeted Therapy Group and Drug Development Unit, The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, SM2 5PT Sutton, Surrey, UK.
| |
Collapse
|
665
|
Doctor SM, Tsao CK, Godbold JH, Galsky MD, Oh WK. Is prostate cancer changing?: evolving patterns of metastatic castration-resistant prostate cancer. Cancer 2013; 120:833-9. [PMID: 25302607 DOI: 10.1002/cncr.28494] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/01/2013] [Accepted: 10/04/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) most commonly metastasizes to the bone, and less commonly to nonosseous sites (eg, lymph nodes, liver, lung). With new therapies extending survival in mCRPC, it was hypothesized that the pattern of metastases is changing over time. The pattern of metastatic disease was evaluated in men with mCRPC, as reported in baseline characteristics of prospective clinical trials over 2 decades. METHODS This study identified all phase 2 and 3 therapeutic studies in men with mCRPC in PubMed and American Society of Clinical Oncology abstracts from 1990 to 2012. Studies were excluded if they did not report demographic data and sites of metastasis, or excluded patients with a specific site of metastatic disease (except brain). For each type of metastasis, weighted least squares linear regression models were used to evaluate temporal trends. RESULTS A total of 290 eligible studies (270 phase 2 studies and 20 phase 3 studies) involving 19,110 patients were identified. Between 1990 and 2012, the rate of nonosseous metastasis increased significantly at 1.6% per year (P < .0001), whereas the rate of osseous metastasis decreased at 0.5% per year (P < .0001). The rate of lymph node metastasis increased at 1.4% per year (P < .0001), but the rate of liver and lung metastasis remained relatively stable. CONCLUSIONS A notable change was found in the pattern of metastasis in patients with mCRPC. Because these evolving patterns may have important implications in treatment selection and prognosis, it is crucial that future clinical trials of patients with mCRPC define patients with a uniform reporting of nonosseous metastasis.
Collapse
Affiliation(s)
- Stephanie M Doctor
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | | |
Collapse
|
666
|
Lin D, Wyatt AW, Xue H, Wang Y, Dong X, Haegert A, Wu R, Brahmbhatt S, Mo F, Jong L, Bell RH, Anderson S, Hurtado-Coll A, Fazli L, Sharma M, Beltran H, Rubin M, Cox M, Gout PW, Morris J, Goldenberg L, Volik SV, Gleave ME, Collins CC, Wang Y. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer Res 2013; 74:1272-83. [PMID: 24356420 DOI: 10.1158/0008-5472.can-13-2921-t] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Standardized and reproducible preclinical models that recapitulate the dynamics of prostate cancer are urgently needed. We established a bank of transplantable patient-derived prostate cancer xenografts that capture the biologic and molecular heterogeneity currently confounding prognostication and therapy development. Xenografts preserved the histopathology, genome architecture, and global gene expression of donor tumors. Moreover, their aggressiveness matched patient observations, and their response to androgen withdrawal correlated with tumor subtype. The panel includes the first xenografts generated from needle biopsy tissue obtained at diagnosis. This advance was exploited to generate independent xenografts from different sites of a primary site, enabling functional dissection of tumor heterogeneity. Prolonged exposure of adenocarcinoma xenografts to androgen withdrawal led to castration-resistant prostate cancer, including the first-in-field model of complete transdifferentiation into lethal neuroendocrine prostate cancer. Further analysis of this model supports the hypothesis that neuroendocrine prostate cancer can evolve directly from adenocarcinoma via an adaptive response and yielded a set of genes potentially involved in neuroendocrine transdifferentiation. We predict that these next-generation models will be transformative for advancing mechanistic understanding of disease progression, response to therapy, and personalized oncology.
Collapse
Affiliation(s)
- Dong Lin
- Authors' Affiliations: Vancouver Prostate Centre; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia; Departments of Experimental Therapeutics and Radiation Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Weill Cornell Cancer Center, Weill Cornell Medical College, New York, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
667
|
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
|
668
|
Concurrent AURKA and MYCN gene amplifications are harbingers of lethal treatment-related neuroendocrine prostate cancer. Neoplasia 2013; 15:1-10. [PMID: 23358695 DOI: 10.1593/neo.121550] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/19/2012] [Accepted: 11/20/2012] [Indexed: 01/12/2023] Open
Abstract
Neuroendocrine prostate cancer (NEPC), also referred to as anaplastic prostate cancer, is a lethal tumor that most commonly arises in late stages of prostate adenocarcinoma (PCA) with predilection to metastasize to visceral organs. In the current study, we explore for evidence that Aurora kinase A (AURKA) and N-myc (MYCN) gene abnormalities are harbingers of treatment-related NEPC (t-NEPC). We studied primary prostate tissue from 15 hormone naïve PCAs, 51 castration-resistant prostate cancers, and 15 metastatic tumors from 72 patients at different stages of disease progression to t-NEPC, some with multiple specimens. Histologic evaluation, immunohistochemistry, and fluorescence in situ hybridization were performed and correlated with clinical variables. AURKA amplification was identified in overall 65% of PCAs (hormone naïve and treated) from patients that developed t-NEPC and in 86% of metastases. Concurrent amplification of MYCN was present in 70% of primary PCAs, 69% of treated PCAs, and 83% of metastases. In contrast, in an unselected PCA cohort, AURKA and MYCN amplifications were identified in only 5% of 169 cases. When metastatic t-NEPC was compared to primary PCA from the same patients, there was 100% concordance of ERG rearrangement, 100% concordance of AURKA amplification, and 60% concordance of MYCN amplification. In tumors with mixed features, there was also 100% concordance of ERG rearrangement and 94% concordance of AURKA and MYCN co-amplification between areas of NEPC and adenocarcinoma. AURKA and MYCN amplifications may be prognostic and predictive biomarkers, as they are harbingers of tumors at risk of progressing to t-NEPC after hormonal therapy.
Collapse
|
669
|
Huang SP, Lin VC, Lee YC, Yu CC, Huang CY, Chang TY, Lee HZ, Juang SH, Lu TL, Bao BY. Genetic variants in nuclear factor-kappa B binding sites are associated with clinical outcomes in prostate cancer patients. Eur J Cancer 2013; 49:3729-37. [DOI: 10.1016/j.ejca.2013.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 06/26/2013] [Accepted: 07/15/2013] [Indexed: 11/16/2022]
|
670
|
Svensson C, Ceder J, Iglesias-Gato D, Chuan YC, Pang ST, Bjartell A, Martinez RM, Bott L, Helczynski L, Ulmert D, Wang Y, Niu Y, Collins C, Flores-Morales A. REST mediates androgen receptor actions on gene repression and predicts early recurrence of prostate cancer. Nucleic Acids Res 2013; 42:999-1015. [PMID: 24163104 PMCID: PMC3902919 DOI: 10.1093/nar/gkt921] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The androgen receptor (AR) is a key regulator of prostate tumorgenesis through actions that are not fully understood. We identified the repressor element (RE)-1 silencing transcription factor (REST) as a mediator of AR actions on gene repression. Chromatin immunoprecipitation showed that AR binds chromatin regions containing well-characterized cis-elements known to mediate REST transcriptional repression, while cell imaging studies confirmed that REST and AR closely co-localize in vivo. Androgen-induced gene repression also involves modulation of REST protein turnover through actions on the ubiquitin ligase β-TRCP. Androgen deprivation or AR blockage with inhibitor MDV3100 (Enzalutamide) leads to neuroendocrine (NE) differentiation, a phenomenon that is mimicked by REST inactivation. Gene expression profiling revealed that REST not only acts to repress neuronal genes but also genes involved in cell cycle progression, including Aurora Kinase A, that has previously been implicated in the growth of NE-like castration-resistant tumors. The analysis of prostate cancer tissue microarrays revealed that tumors with reduced expression of REST have higher probability of early recurrence, independently of their Gleason score. The demonstration that REST modulates AR actions in prostate epithelia and that REST expression is negatively correlated with disease recurrence after prostatectomy, invite a deeper characterization of its role in prostate carcinogenesis.
Collapse
Affiliation(s)
- Charlotte Svensson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark, Division of Urological Cancers, Department of Clinical Sciences, Skåne University Hospital, Lund University, 20502 Malmö, Sweden, Department of Urology, Chang Gung Memorial Hospital, Tao-Yuan 33305, Taiwan, R.O.C., Department of Epidemiology, Karolinska Institutet, 171 77 Stockholm, Sweden, Department of Cell and Molecular Biology, Karolinska Institute, 171 77 Stockholm, Sweden, Regional Laboratories Region Skåne, Clinical Pathology, 205 80 Malmö, Sweden, Department of Surgery (Urology), Memorial Sloan-Kettering Cancer Center, New York, NY 100 65, USA, Vancouver Prostate Centre and The Department of Urologic Sciences, University of British Columbia, Vancouver, BC Canada V6H 3Z6 and Tianjin Institute of Urology, Tianjin Medical University, Tianjin 300 211, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
671
|
Khurana E, Fu Y, Colonna V, Mu XJ, Kang HM, Lappalainen T, Sboner A, Lochovsky L, Chen J, Harmanci A, Das J, Abyzov A, Balasubramanian S, Beal K, Chakravarty D, Challis D, Chen Y, Clarke D, Clarke L, Cunningham F, Evani US, Flicek P, Fragoza R, Garrison E, Gibbs R, Gümüş ZH, Herrero J, Kitabayashi N, Kong Y, Lage K, Liluashvili V, Lipkin SM, MacArthur DG, Marth G, Muzny D, Pers TH, Ritchie GRS, Rosenfeld JA, Sisu C, Wei X, Wilson M, Xue Y, Yu F, Dermitzakis ET, Yu H, Rubin MA, Tyler-Smith C, Gerstein M. Integrative annotation of variants from 1092 humans: application to cancer genomics. Science 2013; 342:1235587. [PMID: 24092746 PMCID: PMC3947637 DOI: 10.1126/science.1235587] [Citation(s) in RCA: 270] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Interpreting variants, especially noncoding ones, in the increasing number of personal genomes is challenging. We used patterns of polymorphisms in functionally annotated regions in 1092 humans to identify deleterious variants; then we experimentally validated candidates. We analyzed both coding and noncoding regions, with the former corroborating the latter. We found regions particularly sensitive to mutations ("ultrasensitive") and variants that are disruptive because of mechanistic effects on transcription-factor binding (that is, "motif-breakers"). We also found variants in regions with higher network centrality tend to be deleterious. Insertions and deletions followed a similar pattern to single-nucleotide variants, with some notable exceptions (e.g., certain deletions and enhancers). On the basis of these patterns, we developed a computational tool (FunSeq), whose application to ~90 cancer genomes reveals nearly a hundred candidate noncoding drivers.
Collapse
Affiliation(s)
- Ekta Khurana
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT 06520, USA
| | - Yao Fu
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
| | - Vincenza Colonna
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Cambridge, CB10 1SA, UK
- Institute of Genetics and Biophysics, National Research Council
(CNR), 80131 Naples, Italy
| | - Xinmeng Jasmine Mu
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
| | - Hyun Min Kang
- Center for Statistical Genetics, Biostatistics, University of
Michigan, Ann Arbor, MI 48109, USA
| | - Tuuli Lappalainen
- Department of Genetic Medicine and Development, University of Geneva
Medical School, 1211 Geneva, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of
Geneva, 1211 Geneva, Switzerland
- Swiss Institute of Bioinformatics, 1211 Geneva, Switzerland
| | - Andrea Sboner
- Institute for Precision Medicine and the Department of Pathology and
Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian
Hospital, New York, NY 10065, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute
for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021,
USA
| | - Lucas Lochovsky
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
| | - Jieming Chen
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Integrated Graduate Program in Physical and Engineering Biology,
Yale University, New Haven, CT 06520, USA
| | - Arif Harmanci
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT 06520, USA
| | - Jishnu Das
- Department of Biological Statistics and Computational Biology,
Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University,
Ithaca, NY 14853, USA
| | - Alexej Abyzov
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT 06520, USA
| | - Suganthi Balasubramanian
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT 06520, USA
| | - Kathryn Beal
- European Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Dimple Chakravarty
- Institute for Precision Medicine and the Department of Pathology and
Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian
Hospital, New York, NY 10065, USA
| | - Daniel Challis
- Baylor College of Medicine, Human Genome Sequencing Center,
Houston, TX 77030, USA
| | - Yuan Chen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Cambridge, CB10 1SA, UK
| | - Declan Clarke
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Laura Clarke
- European Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Fiona Cunningham
- European Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Uday S. Evani
- Baylor College of Medicine, Human Genome Sequencing Center,
Houston, TX 77030, USA
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Robert Fragoza
- Weill Institute for Cell and Molecular Biology, Cornell University,
Ithaca, NY 14853, USA
- Department of Molecular Biology and Genetics, Cornell University,
Ithaca, NY 14853, USA
| | - Erik Garrison
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Richard Gibbs
- Baylor College of Medicine, Human Genome Sequencing Center,
Houston, TX 77030, USA
| | - Zeynep H. Gümüş
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute
for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021,
USA
- Department of Physiology and Biophysics, Weill Cornell Medical
College, New York, NY, 10065, USA
| | - Javier Herrero
- European Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Naoki Kitabayashi
- Institute for Precision Medicine and the Department of Pathology and
Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian
Hospital, New York, NY 10065, USA
| | - Yong Kong
- Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT 06520, USA
- Keck Biotechnology Resource Laboratory, Yale University, New Haven,
CT 06511, USA
| | - Kasper Lage
- Pediatric Surgical Research Laboratories, MassGeneral Hospital for
Children, Massachusetts General Hospital, Boston, MA 02114, USA
- Analytical and Translational Genetics Unit, Massachusetts General
Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Center for Biological Sequence Analysis, Department of Systems
Biology, Technical University of Denmark, Lyngby, Denmark
- Center for Protein Research, University of Copenhagen, Copenhagen,
Denmark
| | - Vaja Liluashvili
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute
for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021,
USA
- Department of Physiology and Biophysics, Weill Cornell Medical
College, New York, NY, 10065, USA
| | - Steven M. Lipkin
- Department of Medicine, Weill Cornell Medical College, New York, NY
10065, USA
| | - Daniel G. MacArthur
- Analytical and Translational Genetics Unit, Massachusetts General
Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of
Harvard and Massachusetts Institute of Technology (MIT), Cambridge, MA 02142,
USA
| | - Gabor Marth
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Donna Muzny
- Baylor College of Medicine, Human Genome Sequencing Center,
Houston, TX 77030, USA
| | - Tune H. Pers
- Center for Biological Sequence Analysis, Department of Systems
Biology, Technical University of Denmark, Lyngby, Denmark
- Division of Endocrinology and Center for Basic and Translational
Obesity Research, Children’s Hospital, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Graham R. S. Ritchie
- European Molecular Biology Laboratory, European Bioinformatics
Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Jeffrey A. Rosenfeld
- Department of Medicine, Rutgers New Jersey Medical School, Newark,
NJ 07101, USA
- IST/High Performance and Research Computing, Rutgers University
Newark, NJ 07101, USA
- Sackler Institute for Comparative Genomics, American Museum of
Natural History, New York, NY 10024, USA
| | - Cristina Sisu
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT 06520, USA
| | - Xiaomu Wei
- Weill Institute for Cell and Molecular Biology, Cornell University,
Ithaca, NY 14853, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY
10065, USA
| | - Michael Wilson
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Yali Xue
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Cambridge, CB10 1SA, UK
| | - Fuli Yu
- Baylor College of Medicine, Human Genome Sequencing Center,
Houston, TX 77030, USA
| | | | - Emmanouil T. Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva
Medical School, 1211 Geneva, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of
Geneva, 1211 Geneva, Switzerland
- Swiss Institute of Bioinformatics, 1211 Geneva, Switzerland
| | - Haiyuan Yu
- Department of Biological Statistics and Computational Biology,
Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University,
Ithaca, NY 14853, USA
| | - Mark A. Rubin
- Institute for Precision Medicine and the Department of Pathology and
Laboratory Medicine, Weill Cornell Medical College and New York-Presbyterian
Hospital, New York, NY 10065, USA
| | - Chris Tyler-Smith
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Cambridge, CB10 1SA, UK
| | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale
University, New Haven, CT 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT 06520, USA
- Department of Computer Science, Yale University, New Haven, CT
06520, USA
| |
Collapse
|
672
|
Molecular pathology of prostate cancer revealed by next-generation sequencing: opportunities for genome-based personalized therapy. Curr Opin Urol 2013; 23:189-93. [PMID: 23385974 DOI: 10.1097/mou.0b013e32835e9ef4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW This article reviews recently identified genomic mutations in prostate cancer. RECENT FINDINGS Advanced sequencing technologies have made it possible to obtain large amounts of data on genomes and transcriptomes of cancers. Such technologies have been used to sequence prostate cancer of different stages, from treatment-naive cancers, to advanced, castration-resistant cancers to the aggressive small cell neuroendocrine carcinomas. For each category of prostate cancer, distinct and overlapping DNA sequence alterations were discovered, including point mutations, small insertions or deletions, copy number changes and chromosomal rearrangements. There appears to be a stepwise increase in genomic alterations from low risk to high risk to advanced cancers. SUMMARY These novel findings have significantly increased our knowledge of the genetic basis of human prostate cancer and the molecular mechanisms responsible for disease progression and treatment resistance. Some of the lesions are potential therapeutic targets. Studies along this direction will eventually make it possible to design personalized management plans for individual patients.
Collapse
|
673
|
Epigenomic alterations in localized and advanced prostate cancer. Neoplasia 2013; 15:373-83. [PMID: 23555183 DOI: 10.1593/neo.122146] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/28/2013] [Accepted: 01/29/2013] [Indexed: 12/20/2022] Open
Abstract
Although prostate cancer (PCa) is the second leading cause of cancer death among men worldwide, not all men diagnosed with PCa will die from the disease. A critical challenge, therefore, is to distinguish indolent PCa from more advanced forms to guide appropriate treatment decisions. We used Enhanced Reduced Representation Bisulfite Sequencing, a genome-wide high-coverage single-base resolution DNA methylation method to profile seven localized PCa samples, seven matched benign prostate tissues, and six aggressive castration-resistant prostate cancer (CRPC) samples. We integrated these data with RNA-seq and whole-genome DNA-seq data to comprehensively characterize the PCa methylome, detect changes associated with disease progression, and identify novel candidate prognostic biomarkers. Our analyses revealed the correlation of cytosine guanine dinucleotide island (CGI)-specific hypermethylation with disease severity and association of certain breakpoints (deletion, tandem duplications, and interchromosomal translocations) with DNA methylation. Furthermore, integrative analysis of methylation and single-nucleotide polymorphisms (SNPs) uncovered widespread allele-specific methylation (ASM) for the first time in PCa. We found that most DNA methylation changes occurred in the context of ASM, suggesting that variations in tumor epigenetic landscape of individuals are partly mediated by genetic differences, which may affect PCa disease progression. We further selected a panel of 13 CGIs demonstrating increased DNA methylation with disease progression and validated this panel in an independent cohort of 20 benign prostate tissues, 16 PCa, and 8 aggressive CRPCs. These results warrant clinical evaluation in larger cohorts to help distinguish indolent PCa from advanced disease.
Collapse
|
674
|
Das K, Tan P. Molecular cytogenetics: recent developments and applications in cancer. Clin Genet 2013; 84:315-25. [DOI: 10.1111/cge.12229] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/02/2013] [Accepted: 07/02/2013] [Indexed: 12/13/2022]
Affiliation(s)
- K Das
- Cancer and Stem Cell Biology; Duke-NUS Graduate Medical School; Singapore Singapore
| | - P Tan
- Cancer and Stem Cell Biology; Duke-NUS Graduate Medical School; Singapore Singapore
- Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine; National University of Singapore; Singapore Singapore
- Genome Institute of Singapore; Singapore Singapore
- Cellular and Molecular Research; National Cancer Centre of Singapore; Singapore Singapore
| |
Collapse
|
675
|
Nakagawa H. Prostate cancer genomics by high-throughput technologies: genome-wide association study and sequencing analysis. Endocr Relat Cancer 2013; 20:R171-81. [PMID: 23625613 DOI: 10.1530/erc-13-0113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PC) is the most common malignancy in males. It is evident that genetic factors at both germline and somatic levels play critical roles in prostate carcinogenesis. Recently, genome-wide association studies (GWAS) by high-throughput genotyping technology have identified more than 70 germline variants of various genes or chromosome loci that are significantly associated with PC susceptibility. They include multiple 8q24 loci, prostate-specific genes, and metabolism-related genes. Somatic alterations in PC genomes have been explored by high-throughput sequencing technologies such as whole-genome sequencing and RNA sequencing, which have identified a variety of androgen-responsive events and fusion transcripts represented by E26 transformation-specific (ETS) gene fusions. Recent innovations in high-throughput genomic technologies have enabled us to analyze PC genomics more comprehensively, more precisely, and on a larger scale in multiple ethnic groups to increase our understanding of PC genomics and biology in germline and somatic studies, which can ultimately lead to personalized medicine for PC diagnosis, prevention, and therapy. However, these data indicate that the PC genome is more complex and heterogeneous than we expected from GWAS and sequencing analyses.
Collapse
Affiliation(s)
- Hidewaki Nakagawa
- Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| |
Collapse
|
676
|
Mazzoleni S, Jachetti E, Morosini S, Grioni M, Piras IS, Pala M, Bulfone A, Freschi M, Bellone M, Galli R. Gene signatures distinguish stage-specific prostate cancer stem cells isolated from transgenic adenocarcinoma of the mouse prostate lesions and predict the malignancy of human tumors. Stem Cells Transl Med 2013; 2:678-89. [PMID: 23884639 DOI: 10.5966/sctm.2013-0041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The relevant social and economic impact of prostate adenocarcinoma, one of the leading causes of death in men, urges critical improvements in knowledge of the pathogenesis and cure of this disease. These can also be achieved by implementing in vitro and in vivo preclinical models by taking advantage of prostate cancer stem cells (PCSCs). The best-characterized mouse model of prostate cancer is the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. TRAMP mice develop a progressive lesion called prostatic intraepithelial neoplasia that evolves into adenocarcinoma (AD) between 24 and 30 weeks of age. ADs often metastasize to lymph nodes, lung, bones, and kidneys. Eventually, approximately 5% of the mice develop an androgen-independent neuroendocrine adenocarcinoma. Here we report the establishment of long-term self-renewing PCSC lines from the different stages of TRAMP progression by application of the neurosphere assay. Stage-specific prostate cell lines were endowed with the critical features expected from malignant bona fide cancer stem cells, namely, self-renewal, multipotency, and tumorigenicity. Notably, transcriptome analysis of stage-specific PCSCs resulted in the generation of well-defined, meaningful gene signatures, which identify distinct stages of human tumor progression. As such, TRAMP-derived PCSCs represent a novel and valuable preclinical model for elucidating the pathogenetic mechanisms leading to prostate adenocarcinoma and for the identification of molecular mediators to be pursued as therapeutic targets.
Collapse
|
677
|
Sluka P, Davis ID. Cell mates: paracrine and stromal targets for prostate cancer therapy. Nat Rev Urol 2013; 10:441-51. [PMID: 23857181 DOI: 10.1038/nrurol.2013.146] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
After many years of limited treatment options for patients with metastatic castration-resistant prostate cancer (mCRPC), multiple systemic therapies are now available, providing patients with significant improvements in survival, symptom control and bone health. Most of the recent advances in this area have been based on better understanding of mCRPC biology, particularly with respect to the key role of androgen receptor signalling. However, most therapies are targeted towards the malignant epithelial cell component of the cancer and it should not be forgotten that cancer cells exist in close and symbiotic relationships with other components of the tumour. Paracrine and stromal signals are often critical to the growth of the cancer and represent new potential therapeutic targets that are separate from the malignant epithelial cells. The stroma produces numerous growth factors, including vascular endothelial growth factor family members, platelet-derived growth factors and fibroblast growth factors, which are all critical for tumour growth. Targeting prostate-cancer-associated fibroblasts in order to destroy the physical and functional scaffold of a cancer is also a logical approach. The interaction between prostate cancer and the immune system remains an active topic of basic and clinical research, with cytokines, chemokines and growth factors being potential targets for therapy. The biology of epithelial-mesenchymal transition and of circulating tumour cells might also provide insight into new therapeutic targets.
Collapse
Affiliation(s)
- Pavel Sluka
- Monash University Eastern Health Clinical School, Level 2, 5 Arnold Street, Box Hill, Melbourne, VIC 3128, Australia
| | | |
Collapse
|
678
|
The molecular basis for ethnic variation and histological subtype differences in prostate cancer. SCIENCE CHINA-LIFE SCIENCES 2013; 56:780-7. [PMID: 23852643 PMCID: PMC4078990 DOI: 10.1007/s11427-013-4522-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/01/2013] [Indexed: 01/04/2023]
Abstract
Prostate cancer is a common malignancy among men in Western countries. Recently the morbidity and mortality of prostate cancer increase dramatically in several oriental countries including China. Rapidly evolving technology in molecular biology such as high-throughput sequencing and integrative analysis of genomic and transcriptomic landscapes have enabled the identification of key oncogenic events for prostate cancer initiation, progression and resistance to hormonal therapy. These surging data of prostate cancer genome also provide insights on ethnic variation and the differences in histological subtype of this disease. In this review, differences in the incidence of prostate cancer and the prevalence of main genetic alterations between Asian and Western populations are discussed. We also review the recent findings on the mechanisms underlying neuroendocrine differentiation of prostate cancer and the development of small cell neuroendocrine carcinoma after androgen deprivation therapy.
Collapse
|
679
|
Schelling LA, Williamson SR, Zhang S, Yao JL, Wang M, Huang J, Montironi R, Lopez-Beltran A, Emerson RE, Idrees MT, Osunkoya AO, Man YG, Maclennan GT, Baldridge LA, Compérat E, Cheng L. Frequent TMPRSS2-ERG rearrangement in prostatic small cell carcinoma detected by fluorescence in situ hybridization: the superiority of fluorescence in situ hybridization over ERG immunohistochemistry. Hum Pathol 2013; 44:2227-33. [PMID: 23850495 DOI: 10.1016/j.humpath.2013.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/29/2013] [Accepted: 05/01/2013] [Indexed: 11/16/2022]
Abstract
Small cell carcinoma of the prostate is both morphologically and immunohistochemically similar to small cell carcinoma of other organs such as the urinary bladder or lung. TMPRSS2-ERG gene fusion appears to be a highly specific alteration in prostatic carcinoma that is frequently shared by small cell carcinoma. In adenocarcinoma, immunohistochemistry for the ERG protein product has been reported to correlate well with the presence of the gene fusion, although in prostatic small cell carcinoma, this relationship is not completely understood. We evaluated 54 cases of small cell carcinoma of the prostate and compared TMPRSS2-ERG gene fusion status by fluorescence in situ hybridization (FISH) to immunohistochemical staining with antibody to ERG. Of 54 cases of prostatic small cell carcinoma, 26 (48%) were positive for TMPRSS2-ERG gene fusion by FISH and 12 (22%) showed overexpression of ERG protein by immunohistochemistry. Of the 26 cases positive by FISH, 11 were also positive for ERG protein by immunohistochemistry. One tumor was positive by immunohistochemistry but negative by FISH. Urinary bladder small cell carcinoma (n = 25) showed negative results by both methods; however, 2 of 14 small cell carcinomas of other organs (lung, head, and neck) showed positive immunohistochemistry but negative FISH. Positive staining for ERG by immunohistochemistry is present in a subset of prostatic small cell carcinomas and correlates with the presence of TMPRSS2-ERG gene fusion. Therefore, it may be useful in confirming prostatic origin when molecular testing is not accessible. However, sensitivity and specificity of ERG immunohistochemistry in small cell carcinoma are decreased compared to FISH.
Collapse
Affiliation(s)
- Lindsay A Schelling
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
680
|
Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L, Jamin Y, Thway K, Robinson SP, Roels F, Witt O, Fischer M, Chesler L, Eilers M. Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell 2013; 24:75-89. [PMID: 23792191 PMCID: PMC4298657 DOI: 10.1016/j.ccr.2013.05.005] [Citation(s) in RCA: 211] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 04/21/2013] [Accepted: 05/08/2013] [Indexed: 01/08/2023]
Abstract
Amplification of MYCN is a driver mutation in a subset of human neuroendocrine tumors, including neuroblastoma. No small molecules that target N-Myc, the protein encoded by MYCN, are clinically available. N-Myc forms a complex with the Aurora-A kinase, which protects N-Myc from proteasomal degradation. Although stabilization of N-Myc does not require the catalytic activity of Aurora-A, we show here that two Aurora-A inhibitors, MLN8054 and MLN8237, disrupt the Aurora-A/N-Myc complex and promote degradation of N-Myc mediated by the Fbxw7 ubiquitin ligase. Disruption of the Aurora-A/N-Myc complex inhibits N-Myc-dependent transcription, correlating with tumor regression and prolonged survival in a mouse model of MYCN-driven neuroblastoma. We conclude that Aurora-A is an accessible target that makes destabilization of N-Myc a viable therapeutic strategy.
Collapse
Affiliation(s)
- Markus Brockmann
- Comprehensive Cancer Center Mainfranken and Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Evon Poon
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, The Royal Marsden NHS Trust, 15 Cotswold Rd. Belmont, Sutton, Surrey SM2 5NG, UK
| | - Teeara Berry
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, The Royal Marsden NHS Trust, 15 Cotswold Rd. Belmont, Sutton, Surrey SM2 5NG, UK
| | - Anne Carstensen
- Comprehensive Cancer Center Mainfranken and Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Hedwig E. Deubzer
- CCU Pediatric Oncology, DKFZ and Department of Pediatrics 3, University Hospital Heidelberg, Germany, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Lukas Rycak
- Institute of Molecular Biology and Tumor Research (IMT), Emil-Mannkopff-Str. 2, 35037 Marburg, Germany
| | - Yann Jamin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, The Royal Marsden NHS Trust, 15 Cotswold Rd. Belmont, Sutton, Surrey SM2 5NG, UK
| | - Khin Thway
- Division of Pathology, The Institute of Cancer Research, The Royal Marsden NHS Trust, 15 Cotswold Rd. Belmont, Sutton, Surrey SM2 5NG, UK
| | - Simon P. Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, The Royal Marsden NHS Trust, 15 Cotswold Rd. Belmont, Sutton, Surrey SM2 5NG, UK
| | - Frederik Roels
- University Children’s Hospital of Cologne, and Cologne Center for Molecular Medicine (CMMC), University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Olaf Witt
- CCU Pediatric Oncology, DKFZ and Department of Pediatrics 3, University Hospital Heidelberg, Germany, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Matthias Fischer
- University Children’s Hospital of Cologne, and Cologne Center for Molecular Medicine (CMMC), University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Louis Chesler
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, The Royal Marsden NHS Trust, 15 Cotswold Rd. Belmont, Sutton, Surrey SM2 5NG, UK
| | - Martin Eilers
- Comprehensive Cancer Center Mainfranken and Theodor Boveri Institute, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
681
|
Potential predictive biomarkers for individualizing treatment for men with castration-resistant prostate cancer. Cancer J 2013; 19:25-33. [PMID: 23337754 DOI: 10.1097/ppo.0b013e31827e0b9c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
With the surge in therapeutic options for men with castration-resistant prostate cancer (CRPC) comes increasingly complicated treatment decision making, highlighting the need for biomarkers that can identify appropriate patients for specific treatments and accurately assess disease response. Predictive biomarkers are factors related to the disease or the host that are associated with improvements in outcomes, such as survival, due to specific therapies. Such biomarkers have become of paramount importance in oncology to maximize the benefits of novel systemic agents while minimizing harm to individual patients and the costs to society. Given the number of newly approved and expensive systemic therapies, including novel hormonal therapies, chemotherapies, immunotherapies, and bone microenvironment-targeting therapies, predictive biomarkers are needed to give physicians a more rational sense of matching the right patient to the right therapy sequence at a given time. There are currently no validated predictive biomarkers in CRPC. We discuss potential predictive biomarkers in men with CRPC and how these may be developed in the context of therapeutic clinical trials.
Collapse
|
682
|
Goldkorn A, Aparicio AM, Quinn DI. Therapeutic windows and opportunity cost cast upon prostate cancer's fatal shore. Ann Oncol 2013; 24:1717-1720. [PMID: 23798673 DOI: 10.1093/annonc/mdt234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023] Open
Affiliation(s)
- A Goldkorn
- Developmental Therapeutics and Genitourinary Cancer Programs, University of Southern California Norris Comprehensive Cancer Center, Los Angeles
| | - A M Aparicio
- Department of Genitourinary Oncology, M. D. Anderson Cancer Center, Houston, USA
| | - D I Quinn
- Developmental Therapeutics and Genitourinary Cancer Programs, University of Southern California Norris Comprehensive Cancer Center, Los Angeles;.
| |
Collapse
|
683
|
Logothetis CJ, Gallick GE, Maity SN, Kim J, Aparicio A, Efstathiou E, Lin SH. Molecular classification of prostate cancer progression: foundation for marker-driven treatment of prostate cancer. Cancer Discov 2013; 3:849-61. [PMID: 23811619 DOI: 10.1158/2159-8290.cd-12-0460] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recently, many therapeutic agents for prostate cancer have been approved that target the androgen receptor and/or the prostate tumor microenvironment. Each of these therapies has modestly increased patient survival. A better understanding of when in the course of prostate cancer progression specific therapies should be applied, and of what biomarkers would indicate when resistance arises, would almost certainly improve survival due to these therapies. Thus, applying the armamentarium of therapeutic agents in the right sequences in the right combination at the right time is a major goal in prostate cancer treatment. For this to occur, an understanding of prostate cancer evolution during progression is required. In this review, we discuss the current understanding of prostate cancer progression, but challenge the prevailing view by proposing a new model of prostate cancer progression, with the goal of improving biologic classification and treatment strategies. We use this model to discuss how integrating clinical and basic understanding of prostate cancer will lead to better implementation of molecularly targeted therapeutics and improve patient survival.
Collapse
Affiliation(s)
- Christopher J Logothetis
- Departments of 1Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
684
|
Wissing MD, van Diest PJ, van der Wall E, Gelderblom H. Antimitotic agents for the treatment of patients with metastatic castrate-resistant prostate cancer. Expert Opin Investig Drugs 2013; 22:635-61. [PMID: 23586879 DOI: 10.1517/13543784.2013.789858] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Metastatic castrate-resistant prostate cancer (mCRPC) is the second deadliest cancer in men. The group of taxanes, which target microtubules of mitotic cells, is currently the only chemotherapy which has proven to increase overall survival in mCRPC patients. Other mitotic inhibitors are being explored for their clinical potential in mCRPC treatment. AREAS COVERED In this review, we summarize recent developments in the application of mitotic inhibitors for mCRPC from a clinical perspective. The four main groups of mitotic inhibitors currently being tested in clinical trials are microtubule-inhibitors, polo-like kinase 1 inhibitors, aurora kinase inhibitors and kinesin-spindle protein inhibitors. Compounds of these groups of inhibitors that are in clinical development for mCRPC are discussed. For this extensive overview, relevant literature was searched in PubMed and retrieved from clinicaltrials.gov and presentations at ASCO/AACR meetings. EXPERT OPINION In general, mitotic inhibitors are clinically well tolerated but exert limited antitumor activity compared to preclinical study results. However, efficacy of mitotic inhibitors is improving, either by personalizing treatment, by introducing more active compounds, by decreasing resistance of cancer cells against mitotic inhibitors or by using mitotic inhibitors in combination therapies.
Collapse
Affiliation(s)
- Michel D Wissing
- Department of Clinical Oncology, Leiden University Medical Center, Albinusdreef 2 K1-62, 2333ZA Leiden, The Netherlands
| | | | | | | |
Collapse
|
685
|
Wyatt AW, Mo F, Wang Y, Collins CC. The diverse heterogeneity of molecular alterations in prostate cancer identified through next-generation sequencing. Asian J Androl 2013; 15:301-8. [PMID: 23503423 PMCID: PMC3739651 DOI: 10.1038/aja.2013.13] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Prostate cancer is a leading cause of global cancer-related death but attempts to improve diagnoses and develop novel therapies have been confounded by significant patient heterogeneity. In recent years, the application of next-generation sequencing to hundreds of prostate tumours has defined novel molecular subtypes and characterized extensive genomic aberration underlying disease initiation and progression. It is now clear that the heterogeneity observed in the clinic is underpinned by a molecular landscape rife with complexity, where genomic rearrangements and rare mutations combine to amplify transcriptomic diversity. This review dissects our current understanding of prostate cancer 'omics', including the sentinel role of copy number variation, the growing spectrum of oncogenic fusion genes, the potential influence of chromothripsis, and breakthroughs in defining mutation-associated subtypes. Increasing evidence suggests that genomic lesions frequently converge on specific cellular functions and signalling pathways, yet recurrent gene aberration appears rare. Therefore, it is critical that we continue to define individual tumour genomes, especially in the context of their expressed transcriptome. Only through improved characterisation of tumour to tumour variability can we advance to an age of precision therapy and personalized oncology.
Collapse
Affiliation(s)
- Alexander W Wyatt
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| | | | | | | |
Collapse
|
686
|
Kim W, Febbo PG. The current and future role of sequence-based analysis in prostate cancer treatment. Per Med 2013; 10:257-273. [PMID: 29768744 DOI: 10.2217/pme.13.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prostate cancer is the most commonly diagnosed, nondermatologic malignancy in US men. Localized disease can be managed through active surveillance or curative, locally directed therapies, but 30% of men treated with surgery or radiation will need additional (often systemic) treatment for relapsed disease. While spectacular advances in medical treatment of advanced prostate cancer have improved the quality and duration of patients' lives, metastatic prostate cancer remains an incurable, lethal disease that requires additional therapies and better treatment strategies. The advent of ultra-high-throughput sequencing technology provides an opportunity to comprehensively assess the constellation of genetic and molecular events underlying each patient's tumor, and promises to enhance our ability to deliver specifically tailored personalized treatment to men with prostate cancer. The known biological and clinical heterogeneity of prostate cancer presents both opportunities and challenges to the application and utilization of sequence-based analysis to guide prostate cancer treatment.
Collapse
Affiliation(s)
- Won Kim
- University of California, 1600 Divisadero Street, A717, Box 1711, San Francisco, CA 94115, USA
| | - Phillip G Febbo
- University of California, 1600 Divisadero Street, A717, Box 1711, San Francisco, CA 94115, USA.
| |
Collapse
|
687
|
Patel SJ, Molinolo AA, Gutkind S, Crawford NPS. Germline genetic variation modulates tumor progression and metastasis in a mouse model of neuroendocrine prostate carcinoma. PLoS One 2013; 8:e61848. [PMID: 23620793 PMCID: PMC3631138 DOI: 10.1371/journal.pone.0061848] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/14/2013] [Indexed: 01/03/2023] Open
Abstract
Neuroendocrine (NE) differentiation has gained increased attention as a prostate cancer (PC) prognostic marker. The aim of this study is to determine whether host germline genetic variation influences tumor progression and metastasis in C57BL/6-Tg(TRAMP)8247Ng/J (TRAMP) mouse model of aggressive NEPC. TRAMP mice were crossed to the eight progenitor strains of the Collaborative Cross recombinant inbred panel to address this. Tumor growth and metastasis burden were quantified in heterozygous transgene positive F1 male mice at 30 weeks of age. Compared to wild-type C57BL/6J-Tg(TRAMP)824Ng/J males, TRAMP x CAST/EiJ, TRAMP x NOD/ShiLtJ and TRAMP x NZO/HlLtJ F1 males displayed significant increases in tumor growth. Conversely, TRAMP x WSB/EiJ and TRAMP x PWK/PhJ F1 males displayed significant reductions in tumor growth. Interestingly, despite reduced tumor burden, TRAMP x WSB/EiJ males had an increased nodal metastasis burden. Patterns of distant pulmonary metastasis tended to follow the same patterns as that of local dissemination in each of the strains. All tumors and metastases displayed positive staining for NE markers, synaptophysin, and FOXA2. These experiments conclusively demonstrate that the introduction of germline variation by breeding modulates tumor growth, local metastasis burden, and distant metastasis frequency in this model of NEPC. These strains will be useful as model systems to facilitate the identification of germline modifier genes that promote the development of aggressive forms of PC.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/metabolism
- Biomarkers, Tumor/metabolism
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Crosses, Genetic
- Disease Models, Animal
- Disease Progression
- Epithelium/metabolism
- Epithelium/pathology
- Female
- Genetic Variation
- Genotype
- Germ Cells/pathology
- Humans
- Male
- Mice
- Mice, Inbred Strains
- Mice, Transgenic
- Neoplasm Metastasis
- Phenotype
- Prostate/metabolism
- Prostate/pathology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Survival Analysis
- Tumor Burden
Collapse
Affiliation(s)
- Shashank J. Patel
- Metastasis Genetics Section, Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alfredo A. Molinolo
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Silvio Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nigel P. S. Crawford
- Metastasis Genetics Section, Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
688
|
He W, Zhang MG, Wang XJ, Zhong S, Shao Y, Zhu Y, Shen ZJ. AURKA suppression induces DU145 apoptosis and sensitizes DU145 to docetaxel treatment. Am J Transl Res 2013; 5:359-367. [PMID: 23634246 PMCID: PMC3633978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 03/22/2013] [Indexed: 06/02/2023]
Abstract
The palliative therapy effect by docetaxel for CRPC patients makes it urgent to improve the therapy. It was suggested that PI3K and androgen receptor-directed combination therapy may be effective for prostate cancer (PCa) patients PTEN negative. However, for those patients PTEN positive, the mechanism of anti-apoptosis survival of cancer cells is not yet well defined. Amplification of AURKA has been detected in 5% of PCa. In this work, Du145, a PTEN positive PCa cell model, was employed to investigate the role of aurora kinase a (AURKA) on cell growth. Inhibition of AURKA expression by shRNA markedly reduced prostate cancer cell viability. Furthermore, we demonstrate that AURKA inhibition induced a remarkable downregulation of AKT activity and Bax induction. Moreover, specific inhibition of the activity of AURKA, but not other aurora family members, by small molecular chemical inhibitors induced significant cell killing effects. Notably, AURKA inhibition sensitized prostate cancer cells to docetaxel treatment. Our work suggests that AURKA-directed monotherapy or combination therapy with docetaxel could be a potent treatment for PCa patients in future.
Collapse
Affiliation(s)
- Wei He
- Department of Urology, Ruijin hospital, Shanghai Jiaotong University, school of medicine Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
689
|
Roychowdhury S, Chinnaiyan AM. Advancing precision medicine for prostate cancer through genomics. J Clin Oncol 2013; 31:1866-73. [PMID: 23589550 DOI: 10.1200/jco.2012.45.3662] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Prostate cancer is the most common type of cancer in men and the second leading cause of cancer death in men in the United States. The recent surge of high-throughput sequencing of cancer genomes has supported an expanding molecular classification of prostate cancer. Translation of these basic science studies into clinically valuable biomarkers for diagnosis and prognosis and biomarkers that are predictive for therapy is critical to the development of precision medicine in prostate cancer. We review potential applications aimed at improving screening specificity in prostate cancer and differentiating aggressive versus indolent prostate cancers. Furthermore, we review predictive biomarker candidates involving ETS gene rearrangements, PTEN inactivation, and androgen receptor signaling. These and other putative biomarkers may signify aberrant oncogene pathway activation and provide a rationale for matching patients with molecularly targeted therapies in clinical trials. Lastly, we advocate innovations for clinical trial design to incorporate tumor biopsy and molecular characterization to develop biomarkers and understand mechanisms of resistance.
Collapse
Affiliation(s)
- Sameek Roychowdhury
- University of Michigan Medical School, 1400 E. Medical Center Dr, 5316 CCGC, Ann Arbor, MI 48109-5940, USA
| | | |
Collapse
|
690
|
Liu Q, Russell MR, Shahriari K, Jernigan DL, Lioni MI, Garcia FU, Fatatis A. Interleukin-1β promotes skeletal colonization and progression of metastatic prostate cancer cells with neuroendocrine features. Cancer Res 2013; 73:3297-305. [PMID: 23536554 DOI: 10.1158/0008-5472.can-12-3970] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite the progress made in the early detection and treatment of prostate adenocarcinoma, the metastatic lesions from this tumor are incurable. We used genome-wide expression analysis of human prostate cancer cells with different metastatic behavior in animal models to reveal that bone-tropic phenotypes upregulate three genes encoding for the cytokine interleukin-1β (IL-1β), the chemokine CXCL6 (GCP-2), and the protease inhibitor elafin (PI3). The Oncomine database revealed that these three genes are significantly upregulated in human prostate cancer versus normal tissue and correlate with Gleason scores ≥7. This correlation was further validated for IL-1β by immunodetection in prostate tissue arrays. Our study also shows that the exogenous overexpression of IL-1β in nonmetastatic cancer cells promotes their growth into large skeletal lesions in mice, whereas its knockdown significantly impairs the bone progression of highly metastatic cells. In addition, IL-1β secreted by metastatic cells induced the overexpression of COX-2 (PTGS2) in human bone mesenchymal cells treated with conditioned media from bone metastatic prostate cancer cells. Finally, we inspected human tissue specimens from skeletal metastases and detected prostate cancer cells positive for both IL-1β and synaptophysin while concurrently lacking prostate-specific antigen (PSA, KLK3) expression. Collectively, these findings indicate that IL-1β supports the skeletal colonization and metastatic progression of prostate cancer cells with an acquired neuroendocrine phenotype.
Collapse
Affiliation(s)
- Qingxin Liu
- Department of Pharmacology and Physiology, Pathology and Laboratory Medicine, Drexel University College of Medicine; and Kimmel Cancer Center, Philadelphia, PA 19102, USA
| | | | | | | | | | | | | |
Collapse
|
691
|
|
692
|
Nikonova AS, Astsaturov I, Serebriiskii IG, Dunbrack RL, Golemis EA. Aurora A kinase (AURKA) in normal and pathological cell division. Cell Mol Life Sci 2013; 70:661-87. [PMID: 22864622 PMCID: PMC3607959 DOI: 10.1007/s00018-012-1073-7] [Citation(s) in RCA: 321] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 06/05/2012] [Accepted: 06/21/2012] [Indexed: 12/20/2022]
Abstract
Temporally and spatially controlled activation of the Aurora A kinase (AURKA) regulates centrosome maturation, entry into mitosis, formation and function of the bipolar spindle, and cytokinesis. Genetic amplification and mRNA and protein overexpression of Aurora A are common in many types of solid tumor, and associated with aneuploidy, supernumerary centrosomes, defective mitotic spindles, and resistance to apoptosis. These properties have led Aurora A to be considered a high-value target for development of cancer therapeutics, with multiple agents currently in early-phase clinical trials. More recently, identification of additional, non-mitotic functions and means of activation of Aurora A during interphase neurite elongation and ciliary resorption have significantly expanded our understanding of its function, and may offer insights into the clinical performance of Aurora A inhibitors. Here we review the mitotic and non-mitotic functions of Aurora A, discuss Aurora A regulation in the context of protein structural information, and evaluate progress in understanding and inhibiting Aurora A in cancer.
Collapse
Affiliation(s)
- Anna S. Nikonova
- Program in Developmental Therapeutics, Fox Chase Cancer Center, W406, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Igor Astsaturov
- Program in Developmental Therapeutics, Fox Chase Cancer Center, W406, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Ilya G. Serebriiskii
- Program in Developmental Therapeutics, Fox Chase Cancer Center, W406, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Roland L. Dunbrack
- Program in Developmental Therapeutics, Fox Chase Cancer Center, W406, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Erica A. Golemis
- Program in Developmental Therapeutics, Fox Chase Cancer Center, W406, 333 Cottman Ave., Philadelphia, PA 19111 USA
| |
Collapse
|
693
|
Goodin S, DiPaola RS. Cabozantinib in Prostate Cancer: The Beginning of a Precision Paradigm? J Clin Oncol 2013; 31:401-3. [DOI: 10.1200/jco.2012.46.6185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Susan Goodin
- The Cancer Institute of New Jersey; and University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Robert S. DiPaola
- The Cancer Institute of New Jersey; and University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
694
|
Beltran H, Rubin MA. New strategies in prostate cancer: translating genomics into the clinic. Clin Cancer Res 2012; 19:517-23. [PMID: 23248095 DOI: 10.1158/1078-0432.ccr-12-1452] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
With the rapidly developing use of next-generation sequencing technologies, there has been a surge in our knowledge of the genomic landscape of prostate cancer and a movement toward developing a molecular subclassification system for the disease. With this new understanding comes great clinical potential, both for the development of biomarkers as well as new therapeutic targets. Herein, we highlight the potential clinical use of recent discoveries and how they fit into our current paradigm. We describe the challenges that lie ahead as we move from genomic sequencing toward routine clinical practice and adopt precision cancer care for patients with prostate cancer.
Collapse
Affiliation(s)
- Himisha Beltran
- Weill Cornell Cancer Center Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | |
Collapse
|
695
|
Aparicio A, Logothetis CJ, Maity SN. Understanding the lethal variant of prostate cancer: power of examining extremes. Cancer Discov 2012; 1:466-8. [PMID: 22586648 DOI: 10.1158/2159-8290.cd-11-0259] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Small cell prostate carcinoma is a lethal variant of castration-resistant prostate cancer. Beltran and colleagues identified overexpression and amplification of both aurora kinase A (AURKA) and the MYCN proto-oncogene in the small cell prostate carcinomas and propose Aurora kinase A as a potential therapeutic target in this disease subset.
Collapse
Affiliation(s)
- Ana Aparicio
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030-4009, USA
| | | | | |
Collapse
|
696
|
Beltran H, Tagawa ST, Park K, MacDonald T, Milowsky MI, Mosquera JM, Rubin MA, Nanus DM. Challenges in recognizing treatment-related neuroendocrine prostate cancer. J Clin Oncol 2012; 30:e386-9. [PMID: 23169519 DOI: 10.1200/jco.2011.41.5166] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Himisha Beltran
- Weill Cornell Cancer Center; Weill Cornell Medical College, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
697
|
Beltran H, Yelensky R, Frampton GM, Park K, Downing SR, MacDonald TY, Jarosz M, Lipson D, Tagawa ST, Nanus DM, Stephens PJ, Mosquera JM, Cronin MT, Rubin MA. Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. Eur Urol 2012; 63:920-6. [PMID: 22981675 DOI: 10.1016/j.eururo.2012.08.053] [Citation(s) in RCA: 338] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 08/27/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Most personalized cancer care strategies involving DNA sequencing are highly reliant on acquiring sufficient fresh or frozen tissue. It has been challenging to comprehensively evaluate the genome of advanced prostate cancer (PCa) because of limited access to metastatic tissue. OBJECTIVE To demonstrate the feasibility of a novel next-generation sequencing (NGS)-based platform that can be used with archival formalin-fixed paraffin-embedded (FFPE) biopsy tissue to evaluate the spectrum of DNA alterations seen in advanced PCa. DESIGN, SETTING, AND PARTICIPANTS FFPE samples (including archival prostatectomies and prostate needle biopsies) were obtained from 45 patients representing the spectrum of disease: localized PCa, metastatic hormone-naive PCa, and metastatic castration-resistant PCa (CRPC). We also assessed paired primaries and metastases to understand disease heterogeneity and disease progression. INTERVENTION At least 50 ng of tumor DNA was extracted from FFPE samples and used for hybridization capture and NGS using the Illumina HiSeq 2000 platform. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS A total of 3320 exons of 182 cancer-associated genes and 37 introns of 14 commonly rearranged genes were evaluated for genomic alterations. RESULTS AND LIMITATIONS We obtained an average sequencing depth of >900X. Overall, 44% of CRPCs harbored genomic alterations involving the androgen receptor gene (AR), including AR copy number gain (24% of CRPCs) or AR point mutation (20% of CRPCs). Other recurrent mutations included transmembrane protease, serine 2 gene (TMPRSS2):v-ets erythroblastosis virus E26 oncogene homolog (avian) gene (ERG) fusion (44%); phosphatase and tensin homolog gene (PTEN) loss (44%); tumor protein p53 gene (TP53) mutation (40%); retinoblastoma gene (RB) loss (28%); v-myc myelocytomatosis viral oncogene homolog (avian) gene (MYC) gain (12%); and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit α gene (PIK3CA) mutation (4%). There was a high incidence of genomic alterations involving key genes important for DNA repair, including breast cancer 2, early onset gene (BRCA2) loss (12%) and ataxia telangiectasia mutated gene (ATM) mutations (8%); these alterations are potentially targetable with poly(adenosine diphosphate-ribose)polymerase inhibitors. A novel and actionable rearrangement involving the v-raf murine sarcoma viral oncogene homolog B1 gene (BRAF) was also detected. CONCLUSIONS This first-in-principle study demonstrates the feasibility of performing in-depth DNA analyses using FFPE tissue and brings new insight toward understanding the genomic landscape within advanced PCa.
Collapse
Affiliation(s)
- Himisha Beltran
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
698
|
Meulenbeld HJ, Bleuse JP, Vinci EM, Raymond E, Vitali G, Santoro A, Dogliotti L, Berardi R, Cappuzzo F, Tagawa ST, Sternberg CN, Jannuzzo MG, Mariani M, Petroccione A, de Wit R. Randomized phase II study of danusertib in patients with metastatic castration-resistant prostate cancer after docetaxel failure. BJU Int 2012; 111:44-52. [PMID: 22928785 DOI: 10.1111/j.1464-410x.2012.11404.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To determine the efficacy and toxicity of danusertib (formerly PHA-739358) administered i.v. over two different dosing schedules with equivalent dose intensity in patients with metastatic castration-resistant prostate cancer with progressive disease after docetaxel-based treatment. PATIENTS AND METHODS In this open-label, multicentre phase II trial 88 patients were randomly assigned (1:1 ratio) to receive either danusertib 330 mg/m(2) over 6 h i.v. on days 1, 8 and 15 (arm A, n = 43) or 500 mg/m(2) over 24 h i.v. on days 1 and 15 (arm B, n = 38), every 4 weeks. The primary endpoint chosen for this exploratory study was PSA response rate at 3 months. RESULTS Sixty patients (31/43 in arm A and 29/38 in arm B) were evaluable for the primary endpoint. Median progression-free survival was 12 weeks in both arms. PSA response occurred in one patient in each arm; best overall response was stable disease in eight (18.6%) and 13 (34.2%) patients in arms A and B, respectively. Eleven out of 81 (13.6%) treated patients had stable disease for ≥6 months. Danusertib was generally well tolerated; the most common grade 3 and 4 drug-related adverse event was neutropenia which occurred in 37.2% (arm A) and 15.8% (arm B) of the patients. CONCLUSION Danusertib monotherapy shows minimal efficacy in patients with castration-resistant prostate cancer. Further studies are required to establish specific biomarkers predictive for either response or prolonged disease stabilization.
Collapse
Affiliation(s)
- Hielke J Meulenbeld
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
699
|
Lapuk AV, Wu C, Wyatt AW, McPherson A, McConeghy BJ, Brahmbhatt S, Mo F, Zoubeidi A, Anderson S, Bell RH, Haegert A, Shukin R, Wang Y, Fazli L, Hurtado-Coll A, Jones EC, Hach F, Hormozdiari F, Hajirasouliha I, Boutros PC, Bristow RG, Zhao Y, Marra MA, Fanjul A, Maher CA, Chinnaiyan AM, Rubin MA, Beltran H, Sahinalp SC, Gleave ME, Volik SV, Collins CC. From sequence to molecular pathology, and a mechanism driving the neuroendocrine phenotype in prostate cancer. J Pathol 2012; 227:286-97. [PMID: 22553170 DOI: 10.1002/path.4047] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The current paradigm of cancer care relies on predictive nomograms which integrate detailed histopathology with clinical data. However, when predictions fail, the consequences for patients are often catastrophic, especially in prostate cancer where nomograms influence the decision to therapeutically intervene. We hypothesized that the high dimensional data afforded by massively parallel sequencing (MPS) is not only capable of providing biological insights, but may aid molecular pathology of prostate tumours. We assembled a cohort of six patients with high-risk disease, and performed deep RNA and shallow DNA sequencing in primary tumours and matched metastases where available. Our analysis identified copy number abnormalities, accurately profiled gene expression levels, and detected both differential splicing and expressed fusion genes. We revealed occult and potentially dormant metastases, unambiguously supporting the patients' clinical history, and implicated the REST transcriptional complex in the development of neuroendocrine prostate cancer, validating this finding in a large independent cohort. We massively expand on the number of novel fusion genes described in prostate cancer; provide fresh evidence for the growing link between fusion gene aetiology and gene expression profiles; and show the utility of fusion genes for molecular pathology. Finally, we identified chromothripsis in a patient with chronic prostatitis. Our results provide a strong foundation for further development of MPS-based molecular pathology.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/secondary
- Adenocarcinoma/therapy
- Aged
- Alternative Splicing
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- British Columbia
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cluster Analysis
- Decision Support Techniques
- Gene Dosage
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Gene Fusion
- Genetic Predisposition to Disease
- Humans
- Lymphatic Metastasis
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Neoplasms, Hormone-Dependent/therapy
- Neuroendocrine Cells/metabolism
- Neuroendocrine Cells/pathology
- Nomograms
- Oligonucleotide Array Sequence Analysis
- Patient Selection
- Phenotype
- Precision Medicine
- Prognosis
- Prostate-Specific Antigen/blood
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/therapy
- RNA Interference
- Transfection
Collapse
Affiliation(s)
- Anna V Lapuk
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
700
|
Identification of functionally active, low frequency copy number variants at 15q21.3 and 12q21.31 associated with prostate cancer risk. Proc Natl Acad Sci U S A 2012; 109:6686-91. [PMID: 22496589 DOI: 10.1073/pnas.1117405109] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Copy number variants (CNVs) are a recently recognized class of human germ line polymorphisms and are associated with a variety of human diseases, including cancer. Because of the strong genetic influence on prostate cancer, we sought to identify functionally active CNVs associated with susceptibility of this cancer type. We queried low-frequency biallelic CNVs from 1,903 men of Caucasian origin enrolled in the Tyrol Prostate Specific Antigen Screening Cohort and discovered two CNVs strongly associated with prostate cancer risk. The first risk locus (P = 7.7 × 10(-4), odds ratio = 2.78) maps to 15q21.3 and overlaps a noncoding enhancer element that contains multiple activator protein 1 (AP-1) transcription factor binding sites. Chromosome conformation capture (Hi-C) data suggested direct cis-interactions with distant genes. The second risk locus (P = 2.6 × 10(-3), odds ratio = 4.8) maps to the α-1,3-mannosyl-glycoprotein 4-β-N-acetylglucosaminyltransferase C (MGAT4C) gene on 12q21.31. In vitro cell-line assays found this gene to significantly modulate cell proliferation and migration in both benign and cancer prostate cells. Furthermore, MGAT4C was significantly overexpressed in metastatic versus localized prostate cancer. These two risk associations were replicated in an independent PSA-screened cohort of 800 men (15q21.3, combined P = 0.006; 12q21.31, combined P = 0.026). These findings establish noncoding and coding germ line CNVs as significant risk factors for prostate cancer susceptibility and implicate their role in disease development and progression.
Collapse
|