51
|
Peak TC, Panigrahi GK, Praharaj P, Su Y, Shi L, Chyr J, Rivera-Chávez J, Flores-Bocanegra L, Singh R, Vander Griend DJ, Oberlies NH, Kerr BA, Hemal A, Bitting RL, Deep G. Syntaxin 6-mediated exosome secretion regulates enzalutamide resistance in prostate cancer. Mol Carcinog 2020; 59:62-72. [PMID: 31674708 PMCID: PMC6916724 DOI: 10.1002/mc.23129] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) deaths are typically the result of metastatic castration-resistant PCa (mCRPC). Recently, enzalutamide (Enz), an oral androgen receptor inhibitor, was approved for treating patients with mCRPC. Invariably, all PCa patients eventually develop resistance against Enz. Therefore, novel strategies aimed at overcoming Enz resistance are needed to improve the survival of PCa patients. The role of exosomes in drug resistance has not been fully elucidated in PCa. Therefore, we set out to better understand the exosome's role in the mechanism underlying Enz-resistant PCa. Results showed that Enz-resistant PCa cells (C4-2B, CWR-R1, and LNCaP) secreted significantly higher amounts of exosomes (2-4 folds) compared to Enz-sensitive counterparts. Inhibition of exosome biogenesis in resistant cells by GW4869 and dimethyl amiloride strongly decreased their cell viability. Mechanistic studies revealed upregulation of syntaxin 6 as well as its increased colocalization with CD63 in Enz-resistant PCa cells compared to Enz-sensitive cells. Syntaxin 6 knockdown by specific small interfering RNAs in Enz-resistant PCa cells (C4-2B and CWR-R1) resulted in reduced cell number and increased cell death in the presence of Enz. Furthermore, syntaxin 6 knockdown significantly reduced the exosome secretion in both Enz-resistant C4-2B and CWR-R1 cells. The Cancer Genome Atlas analysis showed increased syntaxin 6 expressions associated with higher Gleason score and decreased progression-free survival in PCa patients. Importantly, IHC analysis showed higher syntaxin 6 expression in cancer tissues from Enz-treated patients compared to Enz naïve patients. Overall, syntaxin 6 plays an important role in the secretion of exosomes and increased survival of Enz-resistant PCa cells.
Collapse
Affiliation(s)
- Taylor C. Peak
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Gati K Panigrahi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Prakash Praharaj
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Yixin Su
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Lihong Shi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jacqueline Chyr
- School of Bioinformatics, University of Texas Health Science Center, Houston, Texas
| | - José Rivera-Chávez
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro; Greensboro, North Carolina
| | - Laura Flores-Bocanegra
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro; Greensboro, North Carolina
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - Nicholas H. Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro; Greensboro, North Carolina
| | - Bethany A. Kerr
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Wake Forest Baptist Comprehensive Cancer Center
- Department of Urology
| | | | - Rhonda L. Bitting
- Internal Medicine-Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Wake Forest Baptist Comprehensive Cancer Center
- Department of Urology
| |
Collapse
|
52
|
Cao Q, Song Z, Ruan H, Wang C, Yang X, Bao L, Wang K, Cheng G, Xu T, Xiao W, Xiong Z, Liu D, Yang M, Zhou D, Yang H, Chen K, Zhang X. Targeting the KIF4A/AR Axis to Reverse Endocrine Therapy Resistance in Castration-resistant Prostate Cancer. Clin Cancer Res 2019; 26:1516-1528. [PMID: 31796514 DOI: 10.1158/1078-0432.ccr-19-0396] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/25/2019] [Accepted: 11/26/2019] [Indexed: 11/16/2022]
MESH Headings
- Aged
- Aged, 80 and over
- Androgen Receptor Antagonists/pharmacology
- Animals
- Benzamides
- Cell Line, Tumor
- Cell Proliferation
- Databases, Genetic/statistics & numerical data
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Humans
- Kinesins/antagonists & inhibitors
- Kinesins/metabolism
- Male
- Mice
- Mice, Nude
- Middle Aged
- Nitriles
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/pharmacology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Androgen/chemistry
- Receptors, Androgen/metabolism
- Survival Rate
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Qi Cao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengshuai Song
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Yang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Bao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - TianBo Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Yang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Diwei Zhou
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China.
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Insititute of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
53
|
Udhane V, Maranto C, Hoang DT, Gu L, Erickson A, Devi S, Talati PG, Banerjee A, Iczkowski KA, Jacobsohn K, See WA, Mirtti T, Kilari D, Nevalainen MT. Enzalutamide-Induced Feed-Forward Signaling Loop Promotes Therapy-Resistant Prostate Cancer Growth Providing an Exploitable Molecular Target for Jak2 Inhibitors. Mol Cancer Ther 2019; 19:231-246. [PMID: 31548294 DOI: 10.1158/1535-7163.mct-19-0508] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/16/2019] [Accepted: 09/17/2019] [Indexed: 01/03/2023]
Abstract
The second-generation antiandrogen, enzalutamide, is approved for castrate-resistant prostate cancer (CRPC) and targets androgen receptor (AR) activity in CRPC. Despite initial clinical activity, acquired resistance to enzalutamide arises rapidly and most patients develop terminal disease. Previous work has established Stat5 as a potent inducer of prostate cancer growth. Here, we investigated the significance of Jak2-Stat5 signaling in resistance of prostate cancer to enzalutamide. The levels of Jak2 and Stat5 mRNA, proteins and activation were evaluated in prostate cancer cells, xenograft tumors, and clinical prostate cancers before and after enzalutamide therapy. Jak2 and Stat5 were suppressed by genetic knockdown using lentiviral shRNA or pharmacologic inhibitors. Responsiveness of primary and enzalutamide-resistant prostate cancer to pharmacologic inhibitors of Jak2-Stat5 signaling was assessed in vivo in mice bearing prostate cancer xenograft tumors. Patient-derived prostate cancers were tested for responsiveness to Stat5 blockade as second-line treatment after enzalutamide ex vivo in tumor explant cultures. Enzalutamide-liganded AR induces sustained Jak2-Stat5 phosphorylation in prostate cancer leading to the formation of a positive feed-forward loop, where activated Stat5, in turn, induces Jak2 mRNA and protein levels contributing to further Jak2 activation. Mechanistically, enzalutamide-liganded AR induced Jak2 phosphorylation through a process involving Jak2-specific phosphatases. Stat5 promoted prostate cancer growth during enzalutamide treatment. Jak2-Stat5 inhibition induced death of prostate cancer cells and patient-derived prostate cancers surviving enzalutamide treatment and blocked enzalutamide-resistant tumor growth in mice. This work introduces a novel concept of a pivotal role of hyperactivated Jak2-Stat5 signaling in enzalutamide-resistant prostate cancer, which is readily targetable by Jak2 inhibitors in clinical development.
Collapse
Affiliation(s)
- Vindhya Udhane
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Pharmacology and Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Cristina Maranto
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Pharmacology and Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David T Hoang
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Lei Gu
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew Erickson
- Department of Pathology, Medicum, University of Helsinki, Helsinki, Finland.,Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Savita Devi
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Pharmacology and Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Pooja G Talati
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Anjishnu Banerjee
- Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kenneth Jacobsohn
- Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Urology and Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - William A See
- Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Urology and Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Tuomas Mirtti
- Department of Pathology, Medicum, University of Helsinki, Helsinki, Finland.,Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland.,Department of Pathology, HUSLAB and Helsinki University Hospital, Helsinki, Finland
| | - Deepak Kilari
- Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin and Milwaukee VA Medical Center, Milwaukee, Wisconsin
| | - Marja T Nevalainen
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin. .,Department of Pharmacology and Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Prostate Cancer Center of Excellence at Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
54
|
Kothari V, Goodwin JF, Zhao SG, Drake JM, Yin Y, Chang SL, Evans JR, Wilder-Romans K, Gabbara K, Dylgjeri E, Chou J, Sun G, Tomlins SA, Mehra R, Hege K, Filvaroff EH, Schaeffer EM, Karnes RJ, Quigley DA, Rathkopf DE, He HH, Speers C, Spratt DE, Gilbert LA, Ashworth A, Chinnaiyan AM, Raj GV, Knudsen KE, Feng FY. DNA-Dependent Protein Kinase Drives Prostate Cancer Progression through Transcriptional Regulation of the Wnt Signaling Pathway. Clin Cancer Res 2019; 25:5608-5622. [PMID: 31266829 PMCID: PMC6744969 DOI: 10.1158/1078-0432.ccr-18-2387] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/07/2019] [Accepted: 06/20/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Protein kinases are known to play a prominent role in oncogenic progression across multiple cancer subtypes, yet their role in prostate cancer progression remains underexplored. The purpose of this study was to identify kinases that drive prostate cancer progression.Experimental Design: To discover kinases that drive prostate cancer progression, we investigated the association between gene expression of all known kinases and long-term clinical outcomes in tumor samples from 545 patients with high-risk disease. We evaluated the impact of genetic and pharmacologic inhibition of the most significant kinase associated with metastatic progression in vitro and in vivo. RESULTS DNA-dependent protein kinase (DNAPK) was identified as the most significant kinase associated with metastatic progression in high-risk prostate cancer. Inhibition of DNAPK suppressed the growth of both AR-dependent and AR-independent prostate cancer cells. Gene set enrichment analysis nominated Wnt as the top pathway associated with DNAPK. We found that DNAPK interacts with the Wnt transcription factor LEF1 and is critical for LEF1-mediated transcription. CONCLUSIONS Our data show that DNAPK drives prostate cancer progression through transcriptional regulation of Wnt signaling and is an attractive therapeutic target in aggressive prostate cancer.
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Radiation Oncology, University of California at San Francisco, CA
| | - Jonathan F Goodwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Shuang G Zhao
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Justin M Drake
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Yi Yin
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - S Laura Chang
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Joseph R Evans
- Department of Radiation Oncology, OSF Healthcare, Peoria, Illinois
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Kristina Gabbara
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Emanuela Dylgjeri
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jonathan Chou
- Department of Medicine, University of California at San Francisco, San Francisco, California
| | - Grace Sun
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Scott A Tomlins
- Department of Pathology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Rohit Mehra
- Department of Pathology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | | | | | - Edward M Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
| | | | - Housheng H He
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Luke A Gilbert
- Department of Urology, University of California at San Francisco, San Francisco, California
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Urology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Ganesh V Raj
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Felix Y Feng
- Department of Radiation Oncology, University of California at San Francisco, CA.
- Department of Medicine, University of California at San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
- Department of Urology, University of California at San Francisco, San Francisco, California
| |
Collapse
|
55
|
He Y, Lu J, Ye Z, Hao S, Wang L, Kohli M, Tindall DJ, Li B, Zhu R, Wang L, Huang H. Androgen receptor splice variants bind to constitutively open chromatin and promote abiraterone-resistant growth of prostate cancer. Nucleic Acids Res 2019; 46:1895-1911. [PMID: 29309643 PMCID: PMC5829742 DOI: 10.1093/nar/gkx1306] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/20/2017] [Indexed: 11/13/2022] Open
Abstract
Androgen receptor (AR) splice variants (ARVs) are implicated in development of castration-resistant prostate cancer (CRPC). Upregulation of ARVs often correlates with persistent AR activity after androgen deprivation therapy (ADT). However, the genomic and epigenomic characteristics of ARV-dependent cistrome and the disease relevance of ARV-mediated transcriptome remain elusive. Through integrated chromatin immunoprecipitation coupled sequencing (ChIP-seq) and RNA sequencing (RNA-seq) analysis, we identified ARV-preferential-binding sites (ARV-PBS) and a set of genes preferentially transactivated by ARVs in CRPC cells. ARVs preferentially bind to enhancers located in nucleosome-depleted regions harboring the full AR-response element (AREfull), while full-length AR (ARFL)-PBS are enhancers resided in closed chromatin regions containing the composite FOXA1-nnnn-AREhalf motif. ARV-PBS exclusively overlapped with AR binding sites in castration-resistant (CR) tumors in patients and ARV-preferentially activated genes were up-regulated in abiraterone-resistant patient specimens. Expression of ARV-PBS target genes, such as oncogene RAP2A and cell cycle gene E2F7, were significantly associated with castration resistance, poor survival and tumor progression. We uncover distinct genomic and epigenomic features of ARV-PBS, highlighting that ARVs are useful tools to depict AR-regulated oncogenic genome and epigenome landscapes in prostate cancer. Our data also suggest that the ARV-preferentially activated transcriptional program could be targeted for effective treatment of CRPC.
Collapse
Affiliation(s)
- Yundong He
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Ji Lu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Zhenqing Ye
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Siyuan Hao
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Manish Kohli
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Donald J Tindall
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.,Department of Urology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Benyi Li
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Runzhi Zhu
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA.,Center for Cell Therapy, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Liguo Wang
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.,Department of Urology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.,Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
56
|
Caires-Dos-Santos L, da Silva SV, Smuczek B, de Siqueira AS, Cruz KSP, Barbuto JAM, Augusto TM, Freitas VM, Carvalho HF, Jaeger RG. Laminin-derived peptide C16 regulates Tks expression and reactive oxygen species generation in human prostate cancer cells. J Cell Physiol 2019; 235:587-598. [PMID: 31254281 DOI: 10.1002/jcp.28997] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 01/05/2023]
Abstract
Laminin peptides influence cancer biology. We investigated the role of a laminin-derived peptide C16 regulating invadopodia molecules in human prostate cancer cells (DU145). C16 augmented invadopodia activity of DU145 cells, and stimulated expression Tks4, Tks5, cortactin, and membrane-type matrix metalloproteinase 1. Reactive oxygen species generation is also related to invadopodia formation. This prompted us to address whether C16 would induce reactive oxygen species generation in DU145 cells. Quantitative fluorescence and flow cytometry showed that the peptide C16 increased reactive oxygen species in DU145 cells. Furthermore, significant colocalization between Tks5 and reactive oxygen species was observed in C16-treated cells. Results suggested that the peptide C16 increased Tks5 and reactive oxygen species in prostate cancer cells. The role of C16 increasing Tks and reactive oxygen species are novel findings on invadopodia activity.
Collapse
Affiliation(s)
- Livia Caires-Dos-Santos
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Suély V da Silva
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Basilio Smuczek
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Department of Biology, UNICENTRO State University, Guarapuava, PR, Brazil
| | - Adriane S de Siqueira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,School of Dentistry, Positivo University, Curitiba, PR, Brazil
| | - Karen S P Cruz
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil.,Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, AL, Brazil.,Faculty of Nutrition, Federal University of Alagoas, Maceio, AL, Brazil
| | - José Alexandre M Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Taize M Augusto
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil.,Department of Morphology and Basic Pathology, School of Medicine of Jundiai, Jundiai, SP, Brazil
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Ruy G Jaeger
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
57
|
Blee AM, Huang H. Lineage plasticity-mediated therapy resistance in prostate cancer. Asian J Androl 2019; 21:241-248. [PMID: 29900883 PMCID: PMC6498731 DOI: 10.4103/aja.aja_41_18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/08/2018] [Indexed: 12/21/2022] Open
Abstract
Therapy resistance is a significant challenge for prostate cancer treatment in clinic. Although targeted therapies such as androgen deprivation and androgen receptor (AR) inhibition are effective initially, tumor cells eventually evade these strategies through multiple mechanisms. Lineage reprogramming in response to hormone therapy represents a key mechanism that is increasingly observed. The studies in this area have revealed specific combinations of alterations present in adenocarcinomas that provide cells with the ability to transdifferentiate and perpetuate AR-independent tumor growth after androgen-based therapies. Interestingly, several master regulators have been identified that drive plasticity, some of which also play key roles during development and differentiation of the cell lineages in the normal prostate. Thus, further study of each AR-independent tumor type and understanding underlying mechanisms are warranted to develop combinational therapies that combat lineage plasticity in prostate cancer.
Collapse
Affiliation(s)
- Alexandra M Blee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
- Biochemistry and Molecular Biology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
- Department of Urology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
58
|
Stelloo S, Bergman AM, Zwart W. Androgen receptor enhancer usage and the chromatin regulatory landscape in human prostate cancers. Endocr Relat Cancer 2019; 26:R267-R285. [PMID: 30865928 DOI: 10.1530/erc-19-0032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
The androgen receptor (AR) is commonly known as a key transcription factor in prostate cancer development, progression and therapy resistance. Genome-wide chromatin association studies revealed that transcriptional regulation by AR mainly depends on binding to distal regulatory enhancer elements that control gene expression through chromatin looping to gene promoters. Changes in the chromatin epigenetic landscape and DNA sequence can locally alter AR-DNA-binding capacity and consequently impact transcriptional output and disease outcome. The vast majority of reports describing AR chromatin interactions have been limited to cell lines, identifying numerous other factors and interacting transcription factors that impact AR chromatin interactions. Do these factors also impact AR cistromics - the genome-wide chromatin-binding landscape of AR - in vivo? Recent technological advances now enable researchers to identify AR chromatin-binding sites and their target genes in human specimens. In this review, we provide an overview of the different factors that influence AR chromatin binding in prostate cancer specimens, which is complemented with knowledge from cell line studies. Finally, we discuss novel perspectives on studying AR cistromics in clinical samples.
Collapse
Affiliation(s)
- Suzan Stelloo
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andries M Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
59
|
Efstathiou E, Titus M, Wen S, Troncoso P, Hoang A, Corn P, Prokhorova I, Araujo J, Dmuchowski C, Melhem-Bertrandt A, Patil S, Logothetis CJ. Enzalutamide in Combination with Abiraterone Acetate in Bone Metastatic Castration-resistant Prostate Cancer Patients. Eur Urol Oncol 2019; 3:119-127. [PMID: 31412017 DOI: 10.1016/j.euo.2019.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 12/30/2018] [Accepted: 01/08/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND It is hypothesised that cotargeting the androgen receptor (AR) and paracrine androgen biosynthesis with enzalutamide and abiraterone acetate in metastatic castration-resistant prostate cancer (mCRPC) will dissipate adaptive feedback loops observed with either agent alone. OBJECTIVE To assess the safety, efficacy, androgen signalling/metabolome, and drug-drug interactions (DDIs) of enzalutamide with abiraterone acetate in progressive bone mCRPC (bmCRPC). DESIGN, SETTING, AND PARTICIPANTS This open-label, single-centre interventional study was conducted in bmCRPC patients. INTERVENTION Enzalutamide 160mg and abiraterone acetate 1000mg once daily; prednisone 5mg twice daily. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Adverse events (AEs), prostate-specific antigen (PSA) response, progression-free survival (PFS), tumour biomarker/metabolite expression, and Cmin plasma concentrations were evaluated. RESULTS AND LIMITATIONS Sixty patients were enrolled. Common AEs independent of grade/causality included fatigue (72%), hyperglycaemia (67%), alkaline phosphatase (ALP) elevation (53%), and hot flush (43%). Grade 3 AEs included hypertension (17%), alanine aminotransferase elevation (12%), ALP elevation (5%), and arthralgia (5%). No treatment-related grade 4 AEs or deaths were reported. Median treatment-discontinuation time was 312d (95% confidence interval [CI] 196.0-483.0). Maximal PSA decline ≥50% and ≥90% occurred in 46 (77%) and 29 (48%) patients, respectively. Median PFS was 251d (95% CI 147-337). At week 9, median tumour microenvironment androgens, precursors, and nuclear AR expression decreased (p<0.001). The baseline tumour AR C/N terminal ratio of ≥80% was associated with treatment benefit. At enzalutamide steady state, abiraterone acetate Cmin was ∼23% lower (range 14.05-200.5ng/ml) than when given alone. CONCLUSIONS Enzalutamide combined with abiraterone acetate has a manageable safety profile, without a meaningful DDI. Both agents are pharmacodynamically active with no feedback. Efficacy findings do not support significant benefit of combined treatment for unselected bmCRPC. PATIENT SUMMARY This is the first study combining enzalutamide plus abiraterone in bone metastatic castration-resistant prostate cancer. Results show that this combination is safe.
Collapse
Affiliation(s)
- Eleni Efstathiou
- Department of Genitourinary Medical Oncology, Stanford Alexander Tissue Derivatives Laboratory, David H. Koch Center for Applied Research of Genitourinary Cancers, Houston, TX, USA; Department of Clinical Therapeutics, University of Athens, Athens, Greece.
| | - Mark Titus
- Department of Genitourinary Medical Oncology, Stanford Alexander Tissue Derivatives Laboratory, David H. Koch Center for Applied Research of Genitourinary Cancers, Houston, TX, USA
| | - Sijin Wen
- Department of Biostatistics, West Virginia University, Morgantown, WV, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anh Hoang
- Department of Genitourinary Medical Oncology, Stanford Alexander Tissue Derivatives Laboratory, David H. Koch Center for Applied Research of Genitourinary Cancers, Houston, TX, USA
| | - Paul Corn
- Department of Genitourinary Medical Oncology, Stanford Alexander Tissue Derivatives Laboratory, David H. Koch Center for Applied Research of Genitourinary Cancers, Houston, TX, USA
| | - Ina Prokhorova
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Araujo
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, Stanford Alexander Tissue Derivatives Laboratory, David H. Koch Center for Applied Research of Genitourinary Cancers, Houston, TX, USA
| |
Collapse
|
60
|
Li S, Fong KW, Gritsina G, Zhang A, Zhao JC, Kim J, Sharp A, Yuan W, Aversa C, Yang XJ, Nelson PS, Feng FY, Chinnaiyan AM, de Bono JS, Morrissey C, Rettig MB, Yu J. Activation of MAPK Signaling by CXCR7 Leads to Enzalutamide Resistance in Prostate Cancer. Cancer Res 2019; 79:2580-2592. [PMID: 30952632 DOI: 10.1158/0008-5472.can-18-2812] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/19/2019] [Accepted: 03/29/2019] [Indexed: 01/16/2023]
Abstract
Castration-resistant prostate cancer (CRPC) that has developed resistance to the new-generation androgen receptor (AR) antagonist enzalutamide is a lethal disease. Transcriptome analysis of multiple prostate cancer models identified CXCR7, an atypical chemokine receptor, as one of the most upregulated genes in enzalutamide-resistant cells. AR directly repressed CXCR7 by binding to an enhancer 110 kb downstream of the gene and expression was restored upon androgen deprivation. We demonstrate that CXCR7 is a critical regulator of prostate cancer sensitivity to enzalutamide and is required for CRPC growth in vitro and in vivo. Elevated CXCR7 activated MAPK/ERK signaling through ligand-independent, but β-arrestin 2-dependent mechanisms. Examination of patient specimens showed that CXCR7 and pERK levels increased significantly from localized prostate cancer to CRPC and further upon enzalutamide resistance. Preclinical studies revealed remarkable efficacies of MAPK/ERK inhibitors in suppressing enzalutamide-resistant prostate cancer. Overall, these results indicate that CXCR7 may serve as a biomarker of resistant disease in patients with prostate cancer and that disruption of CXCR7 signaling may be an effective strategy to overcome resistance. SIGNIFICANCE: These findings identify CXCR7-mediated MAPK activation as a mechanism of resistance to second-generation antiandrogen therapy, highlighting the therapeutic potential of MAPK/ERK inhibitors in CRPC.
Collapse
Affiliation(s)
- Shangze Li
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ka-Wing Fong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Galina Gritsina
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ali Zhang
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jonathan C Zhao
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jung Kim
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Adam Sharp
- Institute of Cancer Research, London, United Kingdom.,Royal Marsden Hospital, London, United Kingdom
| | - Wei Yuan
- Institute of Cancer Research, London, United Kingdom
| | - Caterina Aversa
- Institute of Cancer Research, London, United Kingdom.,Royal Marsden Hospital, London, United Kingdom
| | - Ximing J Yang
- Department of Pathology, Northwestern University, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Felix Y Feng
- Departments of Radiation Oncology, Urology, and Medicine, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Johann S de Bono
- Institute of Cancer Research, London, United Kingdom.,Royal Marsden Hospital, London, United Kingdom
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Matthew B Rettig
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California.,VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois. .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| |
Collapse
|
61
|
Wu M, Xie Y, Cui X, Huang C, Zhang R, He Y, Li X, Liu M, Cen S, Zhou J. Rational drug design for androgen receptor and glucocorticoids receptor dual antagonist. Eur J Med Chem 2019; 166:232-242. [DOI: 10.1016/j.ejmech.2019.01.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023]
|
62
|
Thaper D, Vahid S, Kaur R, Kumar S, Nouruzi S, Bishop JL, Johansson M, Zoubeidi A. Galiellalactone inhibits the STAT3/AR signaling axis and suppresses Enzalutamide-resistant Prostate Cancer. Sci Rep 2018; 8:17307. [PMID: 30470788 PMCID: PMC6251893 DOI: 10.1038/s41598-018-35612-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022] Open
Abstract
Most prostate cancer patients will progress to a castration-resistant state (CRPC) after androgen ablation therapy and despite the development of new potent anti-androgens, like enzalutamide (ENZ), which prolong survival in CRPC, ENZ-resistance (ENZR) rapidly occurs. Re-activation of the androgen receptor (AR) is a major mechanism of resistance. Interrogating our in vivo derived ENZR model, we discovered that transcription factor STAT3 not only displayed increased nuclear localization but also bound to and facilitated AR activity. We observed increased STAT3 S727 phosphorylation in ENZR cells, which has been previously reported to facilitate AR binding. Strikingly, ENZR cells were more sensitive to inhibition with STAT3 DNA-binding inhibitor galiellalactone (GPA500) compared to CRPC cells. Treatment with GPA500 suppressed AR activity and significantly reduced expression of Cyclin D1, thus reducing cell cycle progression into S phase and hindering cell proliferation. In vivo, GPA500 reduced tumor volume and serum PSA in ENZR xenografts. Lastly, the combination of ENZ and GPA500 was additive in the inhibition of AR activity and proliferation in LNCaP and CRPC cells, providing rationale for combination therapy. Overall, these results suggest that STAT3 inhibition is a rational therapeutic approach for ENZR prostate cancer, and could be valuable in CRPC in combination with ENZ.
Collapse
Affiliation(s)
- Daksh Thaper
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | | | - Sahil Kumar
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Shaghayegh Nouruzi
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | | | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, BC, Canada. .,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
63
|
Linder S, van der Poel HG, Bergman AM, Zwart W, Prekovic S. Enzalutamide therapy for advanced prostate cancer: efficacy, resistance and beyond. Endocr Relat Cancer 2018; 26:R31-R52. [PMID: 30382692 PMCID: PMC6215909 DOI: 10.1530/erc-18-0289] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
Abstract
The androgen receptor drives the growth of metastatic castration-resistant prostate cancer. This has led to the development of multiple novel drugs targeting this hormone-regulated transcription factor, such as enzalutamide – a potent androgen receptor antagonist. Despite the plethora of possible treatment options, the absolute survival benefit of each treatment separately is limited to a few months. Therefore, current research efforts are directed to determine the optimal sequence of therapies, discover novel drugs effective in metastatic castration-resistant prostate cancer and define patient subpopulations that ultimately benefit from these treatments. Molecular studies provide evidence on which pathways mediate treatment resistance and may lead to improved treatment for metastatic castration-resistant prostate cancer. This review provides, firstly a concise overview of the clinical development, use and effectiveness of enzalutamide in the treatment of advanced prostate cancer, secondly it describes translational research addressing enzalutamide response vs resistance and lastly highlights novel potential treatment strategies in the enzalutamide-resistant setting.
Collapse
Affiliation(s)
- Simon Linder
- Division of OncogenomicsOncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Division of UrologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andries M Bergman
- Division of Medical OncologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of OncogenomicsOncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Laboratory of Chemical Biology and Institute for Complex Molecular SystemsDepartment of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Stefan Prekovic
- Division of OncogenomicsOncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Correspondence should be addressed to S Prekovic:
| |
Collapse
|
64
|
Wang S, Yang S, Nan C, Wang Y, He Y, Mu H. Expression of Androgen Receptor Variant 7 (AR-V7) in Circulated Tumor Cells and Correlation with Drug Resistance of Prostate Cancer Cells. Med Sci Monit 2018; 24:7051-7056. [PMID: 30284554 PMCID: PMC6183102 DOI: 10.12659/msm.909669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Prostate cancer is a common type of malignant tumor invading the male reproductive-urinary system, which has increasing incidence worldwide. Androgen receptor variant 7 (AR-V7) participates in regulating prostate cancer cell proliferation and gene expression. This study aimed to investigate the expression of AR-V7 in circulated tumor cells (CTCs) in patients with prostate cancer and to assess its correlation with drug sensitivity against enzalutamide or abiraterone. MATERIAL AND METHODS Blood samples of prostate cancer patients were collected for separating CTCs, in which mRNA expression level of full-length AR and AR-V7 was measured to analyze their correlation with enzalutamide or abiraterone resistance. Progression-free survival (PFS) of patients with different AR-V7 expression levels was compared. AR-V7 was overexpressed in transfected prostate cancer cells, and its effects on proliferation were analyzed by clonal formation assay. RESULTS qRT-PCR showed AR-V7 overexpression in a total of 13 patients; 76.92% of these patients developed drug resistance, the distal metastasis of which was significantly higher than that in the group with AR-V7 downregulation, with lower PFS (p<0.01). In cultured prostate cancer cells, AR-V7 upregulation resulted in a significantly higher clonal formation rate than in the control group with enzalutamide-containing medium (p<0.05). CONCLUSIONS In prostate cancer cells, AR-V7 expression is correlated with drug resistance, as AR-V7 upregulation leads to enhanced proliferation potency of cancer cells, indicating unfavorable prognosis of patients.
Collapse
Affiliation(s)
- Shuaibin Wang
- Department of Urology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Sen Yang
- Department of Urology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Cunjin Nan
- Department of Urology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Yijun Wang
- Department of Urology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Youhua He
- Department of Urology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Haiqi Mu
- Department of Urology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| |
Collapse
|
65
|
Shih TC, Liu R, Wu CT, Li X, Xiao W, Deng X, Kiss S, Wang T, Chen XJ, Carney R, Kung HJ, Duan Y, Ghosh PM, Lam KS. Targeting Galectin-1 Impairs Castration-Resistant Prostate Cancer Progression and Invasion. Clin Cancer Res 2018; 24:4319-4331. [PMID: 29666302 PMCID: PMC6125207 DOI: 10.1158/1078-0432.ccr-18-0157] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/22/2018] [Accepted: 04/11/2018] [Indexed: 12/31/2022]
Abstract
Purpose: The majority of patients with prostate cancer who are treated with androgen-deprivation therapy (ADT) will eventually develop fatal metastatic castration-resistant prostate cancer (mCRPC). Currently, there are no effective durable therapies for patients with mCRPC. High expression of galectin-1 (Gal-1) is associated with prostate cancer progression and poor clinical outcome. The role of Gal-1 in tumor progression is largely unknown. Here, we characterized Gal-1 functions and evaluated the therapeutic effects of a newly developed Gal-1 inhibitor, LLS30, in mCRPC.Experimental Design: Cell viability, colony formation, migration, and invasion assays were performed to examine the effects of inhibition of Gal-1 in CRPC cells. We used two human CRPC xenograft models to assess growth-inhibitory effects of LLS30. Genome-wide gene expression analysis was conducted to elucidate the effects of LLS30 on metastatic PC3 cells.Results: Gal-1 was highly expressed in CRPC cells, but not in androgen-sensitive cells. Gal-1 knockdown significantly inhibited CRPC cells' growth, anchorage-independent growth, migration, and invasion through the suppression of androgen receptor (AR) and Akt signaling. LLS30 targets Gal-1 as an allosteric inhibitor and decreases Gal-1-binding affinity to its binding partners. LLS30 showed in vivo efficacy in both AR-positive and AR-negative xenograft models. LLS30 not only can potentiate the antitumor effect of docetaxel to cause complete regression of tumors, but can also effectively inhibit the invasion and metastasis of prostate cancer cells in vivoConclusions: Our study provides evidence that Gal-1 is an important target for mCRPC therapy, and LLS30 is a promising small-molecule compound that can potentially overcome mCRPC. Clin Cancer Res; 24(17); 4319-31. ©2018 AACR.
Collapse
Affiliation(s)
- Tsung-Chieh Shih
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California.
| | - Chun-Te Wu
- Department of Urology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California
| | - Xiaojun Deng
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California
| | - Sophie Kiss
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California
| | - Ting Wang
- Genome Center, University of California, Davis, Davis, California
| | - Xiao-Jia Chen
- Institute of Biomedicine & Cell Biology Department, Jinan University, Guangzhou, China
- National Engineering Research Center of Genetic Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Biotechnological Medicine, Guangdong, Guangzhou, China
| | - Randy Carney
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California
| | - Hsing-Jien Kung
- The Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Yong Duan
- Genome Center, University of California, Davis, Davis, California
| | - Paramita M Ghosh
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California
- Department of Urology, School of Medicine, University of California, Davis, Sacramento, California
- Veterans Affairs Northern California Health Care System-Mather, Mather, California
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California.
- UC Davis NCI-designated Comprehensive Cancer Center, University of California, Davis, Sacramento, California
| |
Collapse
|
66
|
Kong Y, Cheng L, Mao F, Zhang Z, Zhang Y, Farah E, Bosler J, Bai Y, Ahmad N, Kuang S, Li L, Liu X. Inhibition of cholesterol biosynthesis overcomes enzalutamide resistance in castration-resistant prostate cancer (CRPC). J Biol Chem 2018; 293:14328-14341. [PMID: 30089652 DOI: 10.1074/jbc.ra118.004442] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/02/2018] [Indexed: 01/01/2023] Open
Abstract
Enzalutamide, a nonsteroidal second-generation antiandrogen, has been recently approved for the management of castration-resistant prostate cancer (CRPC). Although patients can benefit from enzalutamide at the beginning of this therapy, acquired enzalutamide resistance usually occurs within a short period. This motivated us to investigate the mechanism involved and possible approaches for overcoming enzalutamide resistance in CRPC. In the present study, we found that 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR), a crucial enzyme in the mevalonate pathway for sterol biosynthesis, is elevated in enzalutamide-resistant prostate cancer cell lines. HMGCR knockdown could resensitize these cells to the drug, and HMGCR overexpression conferred resistance to it, suggesting that aberrant HMGCR expression is an important enzalutamide-resistance mechanism in prostate cancer cells. Furthermore, enzalutamide-resistant prostate cancer cells were more sensitive to statins, which are HMGCR inhibitors. Of note, a combination of simvastatin and enzalutamide significantly inhibited the growth of enzalutamide-resistant prostate cancer cells in vitro and tumors in vivo Mechanistically, simvastatin decreased protein levels of the androgen receptor (AR), which was further reduced in combination with enzalutamide. We observed that the decrease in AR may occur through simvastatin-mediated inhibition of the mTOR pathway, whose activation was associated with increased HMGCR and AR expression. These results indicate that simvastatin enhances the efficacy of enzalutamide-based therapy, highlighting the therapeutic potential of statins to overcome enzalutamide resistance in CRPC.
Collapse
Affiliation(s)
- Yifan Kong
- From the Departments of Biochemistry and.,Animal Sciences and
| | - Lijun Cheng
- the Department of Biomedical Informatics, Ohio State University, Columbus, Ohio 43210, and
| | - Fengyi Mao
- From the Departments of Biochemistry and.,Animal Sciences and
| | | | | | - Elia Farah
- From the Departments of Biochemistry and
| | | | | | - Nihal Ahmad
- the Department of Dermatology, University of Wisconsin, Madison, Wisconsin 53715
| | - Shihuan Kuang
- Animal Sciences and.,the Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| | - Lang Li
- the Department of Biomedical Informatics, Ohio State University, Columbus, Ohio 43210, and
| | - Xiaoqi Liu
- From the Departments of Biochemistry and .,the Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
67
|
Regufe da Mota S, Bailey S, Strivens RA, Hayden AL, Douglas LR, Duriez PJ, Borrello MT, Benelkebir H, Ganesan A, Packham G, Crabb SJ. LSD1 inhibition attenuates androgen receptor V7 splice variant activation in castration resistant prostate cancer models. Cancer Cell Int 2018; 18:71. [PMID: 29760584 PMCID: PMC5941811 DOI: 10.1186/s12935-018-0568-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/30/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Castrate resistant prostate cancer (CRPC) is often driven by constitutively active forms of the androgen receptor such as the V7 splice variant (AR-V7) and commonly becomes resistant to established hormonal therapy strategies such as enzalutamide as a result. The lysine demethylase LSD1 is a co-activator of the wild type androgen receptor and a potential therapeutic target in hormone sensitive prostate cancer. We evaluated whether LSD1 could also be therapeutically targeted in CRPC models driven by AR-V7. METHODS We utilised cell line models of castrate resistant prostate cancer through over expression of AR-V7 to test the impact of chemical LSD1 inhibition on AR activation. We validated findings through depletion of LSD1 expression and in prostate cancer cell lines that express AR-V7. RESULTS Chemical inhibition of LSD1 resulted in reduced activation of the androgen receptor through both the wild type and its AR-V7 splice variant forms. This was confirmed and validated in luciferase reporter assays, in LNCaP and 22Rv1 prostate cancer cell lines and in LSD1 depletion experiments. CONCLUSION LSD1 contributes to activation of both the wild type and V7 splice variant forms of the androgen receptor and can be therapeutically targeted in models of CRPC. Further development of this approach is warranted.
Collapse
Affiliation(s)
- Sergio Regufe da Mota
- Cancer Sciences Unit and Cancer Research UK Centre, University of Southampton, Southampton General Hospital, Somers Cancer Research Building, Mailpoint 824, Southampton, SO16 6YD UK
| | - Sarah Bailey
- Cancer Sciences Unit and Cancer Research UK Centre, University of Southampton, Southampton General Hospital, Somers Cancer Research Building, Mailpoint 824, Southampton, SO16 6YD UK
| | - Rosemary A. Strivens
- Cancer Sciences Unit and Cancer Research UK Centre, University of Southampton, Southampton General Hospital, Somers Cancer Research Building, Mailpoint 824, Southampton, SO16 6YD UK
| | - Annette L. Hayden
- Cancer Sciences Unit and Cancer Research UK Centre, University of Southampton, Southampton General Hospital, Somers Cancer Research Building, Mailpoint 824, Southampton, SO16 6YD UK
| | - Leon R. Douglas
- Protein Core Facility, Cancer Research UK and Experimental Cancer Medicine Centres, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD UK
| | - Patrick J. Duriez
- Protein Core Facility, Cancer Research UK and Experimental Cancer Medicine Centres, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD UK
| | | | - Hanae Benelkebir
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ UK
| | - A. Ganesan
- School of Pharmacy, University of East Anglia, Norwich, NR4 7TJ UK
| | - Graham Packham
- Cancer Sciences Unit and Cancer Research UK Centre, University of Southampton, Southampton General Hospital, Somers Cancer Research Building, Mailpoint 824, Southampton, SO16 6YD UK
| | - Simon J. Crabb
- Cancer Sciences Unit and Cancer Research UK Centre, University of Southampton, Southampton General Hospital, Somers Cancer Research Building, Mailpoint 824, Southampton, SO16 6YD UK
| |
Collapse
|
68
|
Wu W, Karelia D, Pramanik K, Amin SG, Sharma AK, Jiang C, Lu J. Phenylbutyl isoselenocyanate induces reactive oxygen species to inhibit androgen receptor and to initiate p53-mediated apoptosis in LNCaP prostate cancer cells. Mol Carcinog 2018; 57:1055-1066. [DOI: 10.1002/mc.22825] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/09/2018] [Accepted: 04/14/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Wei Wu
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Deepkamal Karelia
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Kartick Pramanik
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Shantu G. Amin
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
- Penn State Cancer Institute; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Arun K. Sharma
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
- Penn State Cancer Institute; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Cheng Jiang
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
| | - Junxuan Lu
- Department of Pharmacology; Pennsylvania State College of Medicine; Hershey Pennsylvania
- Penn State Cancer Institute; Pennsylvania State College of Medicine; Hershey Pennsylvania
| |
Collapse
|
69
|
Zhang Z, Cheng L, Li J, Farah E, Atallah NM, Pascuzzi PE, Gupta S, Liu X. Inhibition of the Wnt/β-Catenin Pathway Overcomes Resistance to Enzalutamide in Castration-Resistant Prostate Cancer. Cancer Res 2018; 78:3147-3162. [PMID: 29700003 DOI: 10.1158/0008-5472.can-17-3006] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/07/2018] [Accepted: 04/16/2018] [Indexed: 11/16/2022]
Abstract
Enzalutamide is a second-generation nonsteroidal antiandrogen clinically approved for the treatment of castration-resistant prostate cancer (CRPC), yet resistance to endocrine therapy has limited its success in this setting. Although the androgen receptor (AR) has been associated with therapy failure, the mechanisms underlying this failure have not been elucidated. Bioinformatics analysis predicted that activation of the Wnt/β-catenin pathway and its interaction with AR play a major role in acquisition of enzalutamide resistance. To validate the finding, we show upregulation of β-catenin and AR in enzalutamide-resistant cells, partially due to reduction of β-TrCP-mediated ubiquitination. Although activation of the Wnt/β-catenin pathway in enzalutamide-sensitive cells led to drug resistance, combination of β-catenin inhibitor ICG001 with enzalutamide inhibited expression of stem-like markers, cell proliferation, and tumor growth synergistically in various models. Analysis of clinical datasets revealed a molecule pattern shift in different stages of prostate cancer, where we detected a significant correlation between AR and β-catenin expression. These data identify activation of the Wnt/β-catenin pathway as a major mechanism contributing to enzalutamide resistance and demonstrate the potential to stratify patients with high risk of said resistance.Significance: Wnt/β-catenin inhibition resensitizes prostate cancer cells to enzalutamide. Cancer Res; 78(12); 3147-62. ©2018 AACR.
Collapse
Affiliation(s)
| | - Lijun Cheng
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Jie Li
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Elia Farah
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Nadia M Atallah
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | | | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, Ohio
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana. .,Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|
70
|
Tosco L, Laenen A, Gevaert T, Salmon I, Decaestecker C, Davicioni E, Buerki C, Claessens F, Swinnen J, Goffin K, Oyen R, Everaerts W, Moris L, De Meerleer G, Haustermans K, Joniau S. Neoadjuvant degarelix with or without apalutamide followed by radical prostatectomy for intermediate and high-risk prostate cancer: ARNEO, a randomized, double blind, placebo-controlled trial. BMC Cancer 2018; 18:354. [PMID: 29606109 PMCID: PMC5879743 DOI: 10.1186/s12885-018-4275-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 03/21/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Recent retrospective data suggest that neoadjuvant androgen deprivation therapy can improve the prognosis of high-risk prostate cancer (PCa) patients. Novel androgen receptor pathway inhibitors are nowadays available for treatment of metastatic PCa and these compounds are promising for early stage disease. Apalutamide is a pure androgen antagonist with a very high affinity with the androgen receptor. The combination of apalutamide with degarelix, an LHRH antagonist, could increase the efficacy compared to degarelix alone. OBJECTIVE The primary objective is to assess the difference in proportions of minimal residual disease at prostatectomy specimen between apalutamide + degarelix vs placebo + degarelix. Various secondary endpoints are assessed: variations of different biomarkers at the tumour level (tissue microarrays to evaluate DNA-PKs, PARP, AR and splice variants, PSMA, etc.), whole transcriptome sequencing, exome sequencing and clinical (PSA and testosterone kinetics, early biochemical recurrence free survival, quality of life, safety, etc.) and radiological endpoints. METHODS ARNEO is a single centre, phase II, randomized, double blind, placebo-controlled trial. The plan is to include at least 42 patients per each of the two study arms. Patients with intermediate/high-risk PCa and who are amenable for radical prostatectomy with pelvic lymph node dissection can be included. After signing an informed consent, every patient will undergo a pelvic 68Ga -PSMA-11 PSMA PET/MR and receive degarelix at standard dosage and start assuming apalutamide/placebo (60 mg 4 tablets/day) for 12 weeks. Within thirty days from the last study medication intake the same imaging will be repeated. Every patient will undergo PSA and testosterone testing the day of randomization, before the first drug intake, and after the last dose. Formalin fixed paraffin embedded tumour samples will be collected and used for transcriptome analysis, exome sequencing and immunohistochemistry. DISCUSSION ARNEO will allow us to answer, first, whether the combined treatment can result in an increased proportion of patients with minimal residual disease. Secondly, It will enable the study of the molecular consequences at the level of the tumour. Thirdly, what the consequences are of new generation androgen receptor pathway inhibitors on 68Ga -PSMA-11 PET/MR. Finally, various clinical, safety and quality of life data will be collected. TRIAL REGISTRATION EUDRaCT number: 2016-002854-19 (authorization date 3rd August 2017). clinicalTrial.gov: NCT03080116 .
Collapse
Affiliation(s)
- Lorenzo Tosco
- Urology, Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Annouschka Laenen
- Leuven Biostatistics and Statistical Bioinformatics Center, KU Leuven, Leuven, Belgium
| | - Thomas Gevaert
- Laboratory of Experimental Urology, Organ Systems, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Department of Pathology, AZ Klina, Brasschaat, Belgium
| | - Isabelle Salmon
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Decaestecker
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratories of Image, Signal processing & Acoustics, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | - Frank Claessens
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, Leuven, Belgium
| | - Johan Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
- Leuven Cancer Institute, KU Leuven, University of Leuven, Leuven, Belgium
| | - Karolien Goffin
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, UZ Leuven, Leuven, Belgium
| | - Raymond Oyen
- Department of Radiology Gasthuisberg University Hospitals Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Urology, Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | - Lisa Moris
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, Leuven, Belgium
| | - Gert De Meerleer
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Karin Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Steven Joniau
- Urology, Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
71
|
Prekovic S, Van den Broeck T, Moris L, Smeets E, Claessens F, Joniau S, Helsen C, Attard G. Treatment-induced changes in the androgen receptor axis: Liquid biopsies as diagnostic/prognostic tools for prostate cancer. Mol Cell Endocrinol 2018; 462:56-63. [PMID: 28882555 DOI: 10.1016/j.mce.2017.08.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/26/2017] [Accepted: 08/31/2017] [Indexed: 02/06/2023]
Abstract
Prostate cancer progression and treatment relapse is associated with changes in the androgen receptor axis, and analysis of alternations of androgen receptor signaling is valuable for prognostics and treatment optimization. The profile of androgen receptor axis is currently obtained from biopsy specimens, which are not always easy to obtain. Moreover, the information acquired only provides a snapshot of the tumor biology, with strict spatial and temporal limitations. On the other hand, circulation is easily accessible source of both circulating tumor cells and circulating tumor DNA, which can be sampled at numerous time points. This Review will explore the potential use of androgen receptor axis alternations detectable in the blood in therapeutic decision-making and precision medicine for advancing metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- S Prekovic
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Oncogenomics, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
| | - T Van den Broeck
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Urology, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium
| | - L Moris
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - E Smeets
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - F Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - S Joniau
- Department of Urology, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium
| | - C Helsen
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - G Attard
- The Institute of Cancer Research, London SM2 5NG, UK; Royal Marsden National Health Service Foundation Trust, London SM2 5PT, UK
| |
Collapse
|
72
|
Zhu S, Zhao D, Yan L, Jiang W, Kim JS, Gu B, Liu Q, Wang R, Xia B, Zhao JC, Song G, Mi W, Wang RF, Shi X, Lam HM, Dong X, Yu J, Chen K, Cao Q. BMI1 regulates androgen receptor in prostate cancer independently of the polycomb repressive complex 1. Nat Commun 2018; 9:500. [PMID: 29402932 PMCID: PMC5799368 DOI: 10.1038/s41467-018-02863-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/04/2018] [Indexed: 01/10/2023] Open
Abstract
BMI1, a polycomb group (PcG) protein, plays a critical role in epigenetic regulation of cell differentiation and proliferation, and cancer stem cell self-renewal. BMI1 is upregulated in multiple types of cancer, including prostate cancer. As a key component of polycomb repressive complex 1 (PRC1), BMI1 exerts its oncogenic functions by enhancing the enzymatic activities of RING1B to ubiquitinate histone H2A at lysine 119 and repress gene transcription. Here, we report a PRC1-independent role of BMI1 that is critical for castration-resistant prostate cancer (CRPC) progression. BMI1 binds the androgen receptor (AR) and prevents MDM2-mediated AR protein degradation, resulting in sustained AR signaling in prostate cancer cells. More importantly, we demonstrate that targeting BMI1 effectively inhibits tumor growth of xenografts that have developed resistance to surgical castration and enzalutamide treatment. These results suggest that blocking BMI1 alone or in combination with anti-AR therapy can be more efficient to suppress prostate tumor growth.
Collapse
Affiliation(s)
- Sen Zhu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Dongyu Zhao
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Lin Yan
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Xiangya School of Medicine, Central South University, Changsha, Hunan, 410008, China
| | - Weihua Jiang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Jung-Sun Kim
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Bingnan Gu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Qipeng Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Xiangya School of Medicine, Central South University, Changsha, Hunan, 410008, China
| | - Rui Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Bo Xia
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Jonathan C Zhao
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Gang Song
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing, 100034, China
| | - Wenyi Mi
- Department of Epigenetics and Molecular Carcinogenesis, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Xiaobing Shi
- Department of Epigenetics and Molecular Carcinogenesis, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, WA, 98195, USA.,State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), 999078, China
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Kaifu Chen
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA.
| | - Qi Cao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA. .,Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
| |
Collapse
|
73
|
Overcoming Oncogenic Mediated Tumor Immunity in Prostate Cancer. Int J Mol Sci 2017; 18:ijms18071542. [PMID: 28714919 PMCID: PMC5536030 DOI: 10.3390/ijms18071542] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is being tested intensively in clinical trials for prostate cancer; it includes immune checkpoint inhibition, prostate specific antigen (PSA) vaccines and dendritic cell-based strategies. Despite increasing evidence for clinical responses, the consensus of multiple trials is that prostate cancers are poorly responsive to immunotherapy. Prostate cancer has a high degree of pathological and genetic heterogeneity compared to other cancer types, which may account for immunotherapeutic resistance. This hypothesis also implies that select types of prostate tumors may be differentially responsive to immune-based strategies and that the clinical stage, pathological grade and underlying genetic landscape may be important criteria in identifying tumors that respond to immune therapies. One strategy is to target oncogenic driver pathways in combination with immunotherapies with the goal of overcoming tumor immunity and broadening the number of patients achieving a clinical response. In this analysis, we address the hypothesis that driver oncogenic signaling pathways regulate cancer progression, tumor immunity and resistance to current immune therapeutics in prostate cancer. We propose that increased responsiveness may be achieved through the combined use of immunotherapies and inhibitors targeting tumor cell autonomous pathways that contribute towards anti-tumor immunity in patients with prostate cancer.
Collapse
|
74
|
Li K, Guo Y, Yang X, Zhang Z, Zhang C, Xu Y. ELF5-Mediated AR Activation Regulates Prostate Cancer Progression. Sci Rep 2017; 7:42759. [PMID: 28287091 PMCID: PMC5347131 DOI: 10.1038/srep42759] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/13/2017] [Indexed: 01/31/2023] Open
Abstract
The transcription factor E74-like factor 5 (ELF5) is a potent antioncogene that can prevent epithelial-mesenchymal transition (EMT) and metastasis in prostate cancer (PCa). However, little is known how it suppress the tumor growth and if it can interact with androgen receptor (AR). In this study, we find that the ELF5 is frequently expressed in AR activated PCa cells, where it binds to AR acting as a physiological partner and negatively regulates its transcriptional activity. In addition, the interaction between ELF5 and AR is androgen-dependent. Downregulation of ELF5 by shRNA increases the expression of AR-response genes and the progression of PCa. Moreover, ELF5 is a AR-dependent gene that its expression can be induced by androgen and suppressed by antiandrogen treatment. Notably, forced reduction of ELF5 in LNCaP cells facilitates the binding of AR to ARE in ELF5 gene and enabling its transcription, so that low level ELF5 can turn up its own expression by the negative feedback loop.
Collapse
Affiliation(s)
- Kai Li
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China.,Department of Urology, Tianjin Third Central Hospital, Tianjin 300170, China
| | - Yongmin Guo
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiong Yang
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Zhihong Zhang
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Changwen Zhang
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Yong Xu
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| |
Collapse
|
75
|
Chattopadhyay I, Wang J, Qin M, Gao L, Holtz R, Vessella RL, Leach RW, Gelman IH. Src promotes castration-recurrent prostate cancer through androgen receptor-dependent canonical and non-canonical transcriptional signatures. Oncotarget 2017; 8:10324-10347. [PMID: 28055971 PMCID: PMC5354662 DOI: 10.18632/oncotarget.14401] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
Progression of prostate cancer (PC) to castration-recurrent growth (CRPC) remains dependent on sustained expression and transcriptional activity of the androgen receptor (AR). A major mechanism contributing to CRPC progression is through the direct phosphorylation and activation of AR by Src-family (SFK) and ACK1 tyrosine kinases. However, the AR-dependent transcriptional networks activated by Src during CRPC progression have not been elucidated. Here, we show that activated Src (Src527F) induces androgen-independent growth in human LNCaP cells, concomitant with its ability to induce proliferation/survival genes normally induced by dihydrotestosterone (DHT) in androgen-dependent LNCaP and VCaP cells. Src induces additional gene signatures unique to CRPC cell lines, LNCaP-C4-2 and CWR22Rv1, and to CRPC LuCaP35.1 xenografts. By comparing the Src-induced AR-cistrome and/or transcriptome in LNCaP to those in CRPC and LuCaP35.1 tumors, we identified an 11-gene Src-regulated CRPC signature consisting of AR-dependent, AR binding site (ARBS)-associated genes whose expression is altered by DHT in LNCaP[Src527F] but not in LNCaP cells. The differential expression of a subset (DPP4, BCAT1, CNTNAP4, CDH3) correlates with earlier PC metastasis onset and poorer survival, with the expression of BCAT1 required for Src-induced androgen-independent proliferation. Lastly, Src enhances AR binding to non-canonical ARBS enriched for FOXO1, TOP2B and ZNF217 binding motifs; cooperative AR/TOP2B binding to a non-canonical ARBS was both Src- and DHT-sensitive and correlated with increased levels of Src-induced phosphotyrosyl-TOP2B. These data suggest that CRPC progression is facilitated via Src-induced sensitization of AR to intracrine androgen levels, resulting in the engagement of canonical and non-canonical ARBS-dependent gene signatures.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Binding Sites
- Cell Line, Tumor
- Cell Proliferation
- Dihydrotestosterone/pharmacology
- Disease Progression
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Phosphorylation
- Promoter Regions, Genetic
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/enzymology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Androgen/drug effects
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Signal Transduction
- Time Factors
- Transcription, Genetic/drug effects
- Transcriptome
- Transfection
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Indranil Chattopadhyay
- Department of Life Sciences, School of Basic and Applied Science, Central University of Tamil Nadu, Thiruvarur, Tamil Nadu, India
| | - Jianmin Wang
- Department of Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Maochun Qin
- Department of Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lingqiu Gao
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Renae Holtz
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Robert W. Leach
- Lewis-Sigler Institute for Integrative Genomics, Princeton, NJ, USA
| | - Irwin H. Gelman
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
76
|
Hoang DT, Iczkowski KA, Kilari D, See W, Nevalainen MT. Androgen receptor-dependent and -independent mechanisms driving prostate cancer progression: Opportunities for therapeutic targeting from multiple angles. Oncotarget 2017; 8:3724-3745. [PMID: 27741508 PMCID: PMC5356914 DOI: 10.18632/oncotarget.12554] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/29/2016] [Indexed: 12/25/2022] Open
Abstract
Despite aggressive treatment for localized cancer, prostate cancer (PC) remains a leading cause of cancer-related death for American men due to a subset of patients progressing to lethal and incurable metastatic castrate-resistant prostate cancer (CRPC). Organ-confined PC is treated by surgery or radiation with or without androgen deprivation therapy (ADT), while options for locally advanced and disseminated PC include radiation combined with ADT, or systemic treatments including chemotherapy. Progression to CRPC results from failure of ADT, which targets the androgen receptor (AR) signaling axis and inhibits AR-driven proliferation and survival pathways. The exact mechanisms underlying the transition from androgen-dependent PC to CRPC remain incompletely understood. Reactivation of AR has been shown to occur in CRPC despite depletion of circulating androgens by ADT. At the same time, the presence of AR-negative cell populations in CRPC has also been identified. While AR signaling has been proposed as the primary driver of CRPC, AR-independent signaling pathways may represent additional mechanisms underlying CRPC progression. Identification of new therapeutic strategies to target both AR-positive and AR-negative PC cell populations and, thereby, AR-driven as well as non-AR-driven PC cell growth and survival mechanisms would provide a two-pronged approach to eliminate CRPC cells with potential for synthetic lethality. In this review, we provide an overview of AR-dependent and AR-independent molecular mechanisms which drive CRPC, with special emphasis on the role of the Jak2-Stat5a/b signaling pathway in promoting castrate-resistant growth of PC through both AR-dependent and AR-independent mechanisms.
Collapse
Affiliation(s)
- David T Hoang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Kilari
- Department of Medicine, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - William See
- Department of Urology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marja T Nevalainen
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology/Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
77
|
Combination of sorafenib and enzalutamide as a potential new approach for the treatment of castration-resistant prostate cancer. Cancer Lett 2017; 385:108-116. [DOI: 10.1016/j.canlet.2016.10.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/19/2016] [Accepted: 10/22/2016] [Indexed: 12/12/2022]
|
78
|
Ailanthone targets p23 to overcome MDV3100 resistance in castration-resistant prostate cancer. Nat Commun 2016; 7:13122. [PMID: 27959342 PMCID: PMC5159881 DOI: 10.1038/ncomms13122] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/02/2016] [Indexed: 01/17/2023] Open
Abstract
Androgen receptor (AR) antagonist MDV3100 is the first therapeutic approach in treating castration-resistant prostate cancer (CRPC), but tumours frequently become drug resistant via multiple mechanisms including AR amplification and mutation. Here we identify the small molecule Ailanthone (AIL) as a potent inhibitor of both full-length AR (AR-FL) and constitutively active truncated AR splice variants (AR-Vs). AIL binds to the co-chaperone protein p23 and prevents AR's interaction with HSP90, thus resulting in the disruption of the AR-chaperone complex followed by ubiquitin/proteasome-mediated degradation of AR as well as other p23 clients including AKT and Cdk4, and downregulates AR and its target genes in PCa cell lines and orthotopic animal tumours. In addition, AIL blocks tumour growth and metastasis of CRPC. Finally, AIL possesses favourable drug-like properties such as good bioavailability, high solubility, lack of CYP inhibition and low hepatotoxicity. In general, AIL is a potential candidate for the treatment of CRPC. Prostate cancers often become castration resistant due to alternative expression of androgen receptor (AR) splice variants. Here, the authors screened a library of natural compounds and identified Ailanthone as a potent inhibitor of AR through its binding to the co-chaperone protein p23 that, by preventing AR interaction with HSP90, results in ubiquitin/proteasome-mediated degradation of the receptor.
Collapse
|
79
|
He Y, Wu M, Liu Y, Li Q, Li X, Hu L, Cen S, Zhou J. Identification of Triptophenolide from Tripterygium wilfordii as a Pan-antagonist of Androgen Receptor. ACS Med Chem Lett 2016; 7:1024-1027. [PMID: 27994731 DOI: 10.1021/acsmedchemlett.6b00180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 09/28/2016] [Indexed: 12/21/2022] Open
Abstract
A compound, triptophenolide, derived from Tripterygium wilfordii was identified as an antiandrogen. Triptophenolide inhibits the activity of both wild-type and F876L mutant androgen receptors. Triptophenolide exhibits its antiandrogenic activity through competitive binding with androgen in the hormone-binding pocket, decreasing the expression of androgen receptor, and reducing the nuclear translocation of androgen receptor.
Collapse
Affiliation(s)
- Yang He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Meng Wu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Yangguang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Quanjie Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Xiaoyu Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Laixing Hu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Jinming Zhou
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
80
|
Thomas BC, Kay JD, Menon S, Vowler SL, Dawson SN, Bucklow LJ, Luxton HJ, Johnston T, Massie CE, Pugh M, Warren AY, Barker P, Burling K, Lynch AG, George A, Burge J, Corcoran M, Stearn S, Lamb AD, Sharma NL, Shaw GL, Neal DE, Whitaker HC. Whole blood mRNA in prostate cancer reveals a four-gene androgen regulated panel. Endocr Relat Cancer 2016; 23:797-812. [PMID: 27578825 DOI: 10.1530/erc-16-0287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 08/10/2016] [Indexed: 01/05/2023]
Abstract
Due to increased sensitivity, the expression of circulating nucleotides is rapidly gaining popularity in cancer diagnosis. Whole blood mRNA has been used in studies on a number of cancers, most notably two separate studies that used whole blood mRNA to define non-overlapping signatures of prostate cancer that has become castration independent. Prostate cancer is known to rely on androgens for initial growth, and there is increasing evidence on the importance of the androgen axis in advanced disease. Using whole blood mRNA samples from patients with prostate cancer, we have identified the four-gene panel of FAM129A, MME, KRT7 and SOD2 in circulating mRNA that are differentially expressed in a discovery cohort of metastatic samples. Validation of these genes at the mRNA and protein level was undertaken in additional cohorts defined by risk of relapse following surgery and hormone status. All the four genes were downregulated at the mRNA level in the circulation and in primary tissue, but this was not always reflected in tissue protein expression. MME demonstrated significant differences in the hormone cohorts, whereas FAM129A is downregulated at the mRNA level but is raised at the protein level in tumours. Using published ChIP-seq data, we have demonstrated that this may be due to AR binding at the FAM129A and MME loci in multiple cell lines. These data suggest that whole blood mRNA of androgen-regulated genes has the potential to be used for diagnosis and monitoring of prostate cancer.
Collapse
Affiliation(s)
- Benjamin C Thomas
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Biomarker InitiativeCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Jonathan D Kay
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Biomarker InitiativeCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Molecular Diagnostics and Therapeutics GroupUniversity College London, London, UK
| | - Suraj Menon
- Bioinformatics and Statistics Core FacilityCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Astra Zeneca2 Riverside, Granta Park, Cambridge, UK
| | - Sarah L Vowler
- Bioinformatics and Statistics Core FacilityCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Astra Zeneca2 Riverside, Granta Park, Cambridge, UK
| | - Sarah N Dawson
- Bioinformatics and Statistics Core FacilityCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Laura J Bucklow
- Biomarker InitiativeCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Hayley J Luxton
- Biomarker InitiativeCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Molecular Diagnostics and Therapeutics GroupUniversity College London, London, UK
| | - Thomas Johnston
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Biomarker InitiativeCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Charlie E Massie
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Molecular and Computational Diagnostics GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Michelle Pugh
- Genomics Core FacilityCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Anne Y Warren
- Department of HistopathologyCambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Peter Barker
- National Institute for Health Research Cambridge Biomedical Research Centre Core Biochemistry Assay LaboratoryCambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Keith Burling
- National Institute for Health Research Cambridge Biomedical Research Centre Core Biochemistry Assay LaboratoryCambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Andy G Lynch
- Computational Biology GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Anne George
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Johanna Burge
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Marie Corcoran
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Sara Stearn
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Alastair D Lamb
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Naomi L Sharma
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Greg L Shaw
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK University College Hospital at Westmoreland StreetLondon, UK
| | - David E Neal
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Nuffield Department of Surgical SciencesJohn Radcliffe Hospital, Headington, Oxford, UK
| | - Hayley C Whitaker
- Uro-Oncology Research GroupCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Biomarker InitiativeCancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK Molecular Diagnostics and Therapeutics GroupUniversity College London, London, UK
| |
Collapse
|
81
|
Sarwar M, Semenas J, Miftakhova R, Simoulis A, Robinson B, Wingren AG, Mongan NP, Heery DM, Johnsson H, Abrahamsson PA, Dizeyi N, Luo J, Persson JL. Targeted suppression of AR-V7 using PIP5K1α inhibitor overcomes enzalutamide resistance in prostate cancer cells. Oncotarget 2016; 7:63065-63081. [PMID: 27588408 PMCID: PMC5325347 DOI: 10.18632/oncotarget.11757] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 08/20/2016] [Indexed: 01/05/2023] Open
Abstract
One mechanism of resistance of prostate cancer (PCa) to enzalutamide (MDV3100) treatment is the increased expression of AR variants lacking the ligand binding-domain, the best characterized of which is AR-V7. We have previously reported that Phosphatidylinositol-4-phosphate 5-kinase alpha (PIP5Kα), is a lipid kinase that links to CDK1 and AR pathways. The discovery of PIP5Kα inhibitor highlight the potential of PIP5K1α as a drug target in PCa. In this study, we show that AR-V7 expression positively correlates with PIP5K1α in tumor specimens from PCa patients. Overexpression of AR-V7 increases PIP5K1α, promotes rapid growth of PCa in xenograft mice, whereas inhibition of PIP5K1α by its inhibitor ISA-2011B suppresses the growth and invasiveness of xenograft tumors overexpressing AR-V7. PIP5K1α is a key co-factor for both AR-V7 and AR, which are present as protein-protein complexes predominantly in the nucleus of PCa cells. In addition, PIP5K1α and CDK1 influence AR-V7 expression also through AKT-associated mechanism dependent on PTEN-status. ISA-2011B disrupts protein stabilization of AR-V7 which is dependent on PIP5K1α, leading to suppression of invasive growth of AR-V7-high tumors in xenograft mice. Our study suggests that combination of enzalutamide and PIP5K1α may have a significant impact on refining therapeutic strategies to circumvent resistance to antiandrogen therapies.
Collapse
Affiliation(s)
- Martuza Sarwar
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Julius Semenas
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
- Department of Molecular Biology, Umeå University, Sweden
| | - Regina Miftakhova
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
- Department of Genetics, Kazan Federal University, Kazan, Russia
- Department of Molecular Biology, Umeå University, Sweden
| | - Athanasios Simoulis
- Department of Clinical Pathology and Cytology, Skåne University Hospital, Malmö, Sweden
| | - Brian Robinson
- Department of Pathology, Weill Cornell Medical College, New York, NY, USA
| | - Anette Gjörloff Wingren
- Faculty of Health and Society, Department of Biomedical Science, Malmö University, Malmö, Sweden
| | - Nigel P. Mongan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, United Kingdom
| | - David M. Heery
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Heather Johnsson
- Department of Bio-Diagnosis, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Per-Anders Abrahamsson
- Division of Clinical Urology, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Nishtman Dizeyi
- Division of Clinical Urology, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Jun Luo
- Department of Urology, the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jenny L. Persson
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
- Department of Molecular Biology, Umeå University, Sweden
| |
Collapse
|
82
|
Luk ISU, Shrestha R, Xue H, Wang Y, Zhang F, Lin D, Haegert A, Wu R, Dong X, Collins CC, Zoubeidi A, Gleave ME, Gout PW, Wang Y. BIRC6 Targeting as Potential Therapy for Advanced, Enzalutamide-Resistant Prostate Cancer. Clin Cancer Res 2016; 23:1542-1551. [PMID: 27663589 DOI: 10.1158/1078-0432.ccr-16-0718] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 08/11/2016] [Accepted: 09/05/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Enzalutamide resistance has emerged as a major problem in the management of castration-resistant prostate cancer (CRPC). Research on therapy resistance of CRPCs has primarily focused on the androgen receptor pathway. In contrast, there is limited information on antiapoptotic mechanisms that may facilitate the treatment resistance. The inhibitor of apoptosis proteins (IAP) family is well recognized for its role in promoting treatment resistance of cancers by inhibiting drug-induced apoptosis. Here, we examined whether BIRC6, an IAP family member, has a role in enzalutamide resistance of CRPCs and could provide a therapeutic target for enzalutamide-resistant CRPC.Experimental Design: Use of enzalutamide-resistant CRPC models: (i) the transplantable, first high-fidelity LTL-313BR patient-derived enzalutamide-resistant CRPC tissue xenograft line showing primary enzalutamide resistance, (ii) MR42D and MR49F CRPC cells/xenografts showing acquired enzalutamide resistance. Specific BIRC6 downregulation in these models was produced using a BIRC6-targeting antisense oligonucleotide (ASO-6w2). Gene expression was determined by qRT-PCR and gene expression profiling. Molecular pathways associated with growth inhibition were assessed via gene enrichment analysis.Results: Of eight IAPs examined, BIRC6 was the only one showing elevated expression in both enzalutamide-resistant CRPC models. Treatment with ASO-6w2 markedly suppressed growth of LTL-313BR xenografts and increased tumor apoptosis without inducing major host toxicity. Pathway enrichment analysis indicated that GPCR and matrisome signaling were the most significantly altered pathways. Furthermore, ASO-6w2 inhibited expression of prosurvival genes that were upregulated in the LTL-313BR line.Conclusions:BIRC6 targeting inhibited the growth of enzalutamide-resistant CRPC models and may represent a new option for clinical treatment of advanced, enzalutamide-resistant prostate cancer. Clin Cancer Res; 23(6); 1542-51. ©2016 AACR.
Collapse
Affiliation(s)
- Iris Sze Ue Luk
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Raunak Shrestha
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Hui Xue
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yuwei Wang
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Fang Zhang
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Dong Lin
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Rebecca Wu
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Colin C Collins
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter W Gout
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. .,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
83
|
Graça I, Pereira-Silva E, Henrique R, Packham G, Crabb SJ, Jerónimo C. Epigenetic modulators as therapeutic targets in prostate cancer. Clin Epigenetics 2016; 8:98. [PMID: 27651838 PMCID: PMC5025578 DOI: 10.1186/s13148-016-0264-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/07/2016] [Indexed: 01/24/2023] Open
Abstract
Prostate cancer is one of the most common non-cutaneous malignancies among men worldwide. Epigenetic aberrations, including changes in DNA methylation patterns and/or histone modifications, are key drivers of prostate carcinogenesis. These epigenetic defects might be due to deregulated function and/or expression of the epigenetic machinery, affecting the expression of several important genes. Remarkably, epigenetic modifications are reversible and numerous compounds that target the epigenetic enzymes and regulatory proteins were reported to be effective in cancer growth control. In fact, some of these drugs are already being tested in clinical trials. This review discusses the most important epigenetic alterations in prostate cancer, highlighting the role of epigenetic modulating compounds in pre-clinical and clinical trials as potential therapeutic agents for prostate cancer management.
Collapse
Affiliation(s)
- Inês Graça
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; School of Allied Health Sciences (ESTSP), Polytechnic of Porto, Porto, Portugal
| | - Eva Pereira-Silva
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, The Somers Cancer Research Building, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, S016 6YD UK
| | - Simon J Crabb
- Cancer Research UK Centre, Cancer Sciences, The Somers Cancer Research Building, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, S016 6YD UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
84
|
Chen L, Li J, Farah E, Sarkar S, Ahmad N, Gupta S, Larner J, Liu X. Cotargeting HSP90 and Its Client Proteins for Treatment of Prostate Cancer. Mol Cancer Ther 2016; 15:2107-18. [PMID: 27390342 PMCID: PMC5010925 DOI: 10.1158/1535-7163.mct-16-0241] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/26/2016] [Indexed: 11/16/2022]
Abstract
Castration-resistant prostate cancer (CRPC) is the later stage of prostate cancer when the disease has stopped responding to androgen deprivation therapy (ADT). It has been established that androgen receptor (AR) reactivation is responsible for the recurrence of prostate cancer after ADT. Thus, targeting different pathways that regulate AR stability and activity should be a promising strategy for treatment of CRPC. Heat shock proteins (HSP) are chaperones that modify stability and activity of their client proteins. HSP90, a major player in the HSP family, regulates stability of many proteins, including AR and Polo-like kinase 1 (Plk1), a critical regulator of many cell-cycle events. Further, HSP90 is overexpressed in different cancers, including prostate cancer. Herein, we show that cotreatment of prostate cancer with AR antagonist enzalutamide and HSP90 inhibitor leads to more severe cell death due to a synergistic reduction of AR protein. Interestingly, we show that overexpression of Plk1 rescued the synergistic effect and that cotargeting HSP90 and Plk1 also leads to more severe cell death. Mechanistically, we show that E3 ligase CHIP, in addition to targeting AR, is responsible for the degradation of Plk1 as well. These findings suggest that cotargeting HSP90 and some of its client proteins may be a useful strategy in treatment of CRPC. Mol Cancer Ther; 15(9); 2107-18. ©2016 AACR.
Collapse
Affiliation(s)
- Long Chen
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Jie Li
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Elia Farah
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Sukumar Sarkar
- Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, Ohio
| | - James Larner
- Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana. Center for Cancer Research, Purdue University, West Lafayette, Indiana.
| |
Collapse
|
85
|
Sundahl N, Clarisse D, Bracke M, Offner F, Berghe WV, Beck IM. Selective glucocorticoid receptor-activating adjuvant therapy in cancer treatments. Oncoscience 2016; 3:188-202. [PMID: 27713909 PMCID: PMC5043069 DOI: 10.18632/oncoscience.315] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 07/08/2016] [Indexed: 02/07/2023] Open
Abstract
Although adverse effects and glucocorticoid resistance cripple their chronic use, glucocorticoids form the mainstay therapy for acute and chronic inflammatory disorders, and play an important role in treatment protocols of both lymphoid malignancies and as adjuvant to stimulate therapy tolerability in various solid tumors. Glucocorticoid binding to their designate glucocorticoid receptor (GR), sets off a plethora of cell-specific events including therapeutically desirable effects, such as cell death, as well as undesirable effects, including chemotherapy resistance, systemic side effects and glucocorticoid resistance. In this context, selective GR agonists and modulators (SEGRAMs) with a more restricted GR activity profile have been developed, holding promise for further clinical development in anti-inflammatory and potentially in cancer therapies. Thus far, the research into the prospective benefits of selective GR modulators in cancer therapy limped behind. Our review discusses how selective GR agonists and modulators could improve the therapy regimens for lymphoid malignancies, prostate or breast cancer. We summarize our current knowledge and look forward to where the field should move to in the future. Altogether, our review clarifies novel therapeutic perspectives in cancer modulation via selective GR targeting.
Collapse
Affiliation(s)
- Nora Sundahl
- Laboratory of Experimental Cancer Research (LECR), Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Dorien Clarisse
- Laboratory of Experimental Cancer Research (LECR), Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Receptor Research Laboratories, Nuclear Receptor Lab (NRL), VIB Medical Biotechnology Center, Ghent University, Ghent, Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research (LECR), Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Fritz Offner
- Hematology, Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Ilse M Beck
- Laboratory of Experimental Cancer Research (LECR), Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Gent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
86
|
Rahman HP, Hofland J, Foster PA. In touch with your feminine side: how oestrogen metabolism impacts prostate cancer. Endocr Relat Cancer 2016; 23:R249-66. [PMID: 27194038 DOI: 10.1530/erc-16-0118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 05/18/2016] [Indexed: 12/18/2022]
Abstract
Prostate cancer is the primary cancer in males, with increasing global incidence rates making this malignancy a significant healthcare burden. Androgens not only promote normal prostate maturity but also influence the development and progression of prostate cancer. Intriguingly, evidence now suggests endogenous and exogenous oestrogens, in the form of phytoestrogens, may be equally as relevant as androgens in prostate cancer growth. The prostate gland has the molecular mechanisms, catalysed by steroid sulphatase (STS), to unconjugate and utilise circulating oestrogens. Furthermore, prostate tissue also expresses enzymes essential for local oestrogen metabolism, including aromatase (CYP19A1) and 3β- and 17β-hydroxysteroid dehydrogenases. Increased expression of these enzymes in malignant prostate tissue compared with normal prostate indicates that oestrogen synthesis is favoured in malignancy and thus may influence tumour progression. In contrast to previous reviews, here we comprehensively explore the epidemiological and scientific evidence on how oestrogens impact prostate cancer, particularly focusing on pre-receptor oestrogen metabolism and subsequent molecular action. We analyse how molecular mechanisms and metabolic pathways involved in androgen and oestrogen synthesis intertwine to alter prostate tissue. Furthermore, we speculate on whether oestrogen receptor status in the prostate affects progression of this malignancy.
Collapse
Affiliation(s)
- Habibur P Rahman
- Institute of Metabolism and Systems ResearchUniversity of Birmingham, Birmingham, UK
| | - Johannes Hofland
- Department of Internal MedicineErasmus Medical Center, Rotterdam, The Netherlands
| | - Paul A Foster
- Institute of Metabolism and Systems ResearchUniversity of Birmingham, Birmingham, UK Centre for EndocrinologyDiabetes and Metabolism, Birmingham Healthcare Partners, Birmingham, UK
| |
Collapse
|
87
|
ASC-J9(®) suppresses castration resistant prostate cancer progression via degrading the enzalutamide-induced androgen receptor mutant AR-F876L. Cancer Lett 2016; 379:154-60. [PMID: 27233475 DOI: 10.1016/j.canlet.2016.05.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/27/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022]
Abstract
Androgen deprivation therapy (ADT) with the newly developed powerful anti-androgen enzalutamide (Enz, also known as MDV3100) has promising therapeutic effects to suppress castration resistant prostate cancer (CRPC) and extending patients' lives an extra 4.8 months. However, most Enz therapy eventually fails with the development of Enz resistance. The detailed mechanisms how CRPC develops Enz resistance remain unclear and may involve multiple mechanisms. Among them, the induction of the androgen receptor (AR) mutant AR-F876L in some CRPC patients may represent one driving force that confers Enz resistance. Here, we demonstrate that the AR degradation enhancer, ASC-J9(®), not only degrades wild-type AR, but also has the ability to target AR-F876L. The consequence of suppressing AR-F876L may then abrogate AR-F876L mediated CRPC cell proliferation and metastasis. Thus, developing ASC-J9(®) as a new therapeutic approach may represent a novel therapy to better suppress CRPC that has already developed Enz resistance.
Collapse
|
88
|
Wang L, Wang J, Xiong H, Wu F, Lan T, Zhang Y, Guo X, Wang H, Saleem M, Jiang C, Lu J, Deng Y. Co-targeting hexokinase 2-mediated Warburg effect and ULK1-dependent autophagy suppresses tumor growth of PTEN- and TP53-deficiency-driven castration-resistant prostate cancer. EBioMedicine 2016; 7:50-61. [PMID: 27322458 PMCID: PMC4909365 DOI: 10.1016/j.ebiom.2016.03.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 03/09/2016] [Accepted: 03/16/2016] [Indexed: 01/07/2023] Open
Abstract
Currently, no therapeutic options exist for castration-resistant prostate cancer (CRPC) patients who have developed resistance to the second generation anti-androgen receptor (AR) axis therapy. Here we report that co-deletion of Pten and p53 in murine prostate epithelium, often observed in human CRPC, leads to AR-independent CRPC and thus confers de novo resistance to second generation androgen deprivation therapy (ADT) in multiple independent yet complementary preclinical mouse models. In contrast, mechanism-driven co-targeting hexokinase 2 (HK2)-mediated Warburg effect with 2-deoxyglucose (2-DG) and ULK1-dependent autophagy with chloroquine (CQ) selectively kills cancer cells through intrinsic apoptosis to cause tumor regression in xenograft, leads to a near-complete tumor suppression and remarkably extends survival in Pten-/p53-deficiency-driven CRPC mouse model. Mechanistically, 2-DG causes AMPK phosphorylation, which in turn inhibits mTORC1-S6K1 translation signaling to preferentially block anti-apoptotic protein MCL-l synthesis to prime mitochondria-dependent apoptosis while simultaneously activates ULK1-driven autophagy for cell survival to counteract the apoptotic action of anti-Warburg effect. Accordingly, inhibition of autophagy with CQ sensitizes cancer cells to apoptosis upon 2-DG challenge. Given that 2-DG is recommended for phase II clinical trials for prostate cancer and CQ has been clinically used as an anti-malaria drug for many decades, the preclinical results from our proof-of-principle studies in vivo are imminently translatable to clinical trials to evaluate the therapeutic efficacy by the combination modality for a subset of currently incurable CRPC harboring PTEN and TP53 mutations.
Collapse
Affiliation(s)
- Lei Wang
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Ji Wang
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Hua Xiong
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Fengxia Wu
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Tian Lan
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Yingjie Zhang
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Xiaolan Guo
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Huanan Wang
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Mohammad Saleem
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA
| | - Cheng Jiang
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Junxuan Lu
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Yibin Deng
- Laboratory of Cancer Genetics, The University of Minnesota Hormel Institute, Austin, MN, 55912, USA.
| |
Collapse
|
89
|
Nadal R, Bellmunt J. The evolving role of enzalutamide on the treatment of prostate cancer. Future Oncol 2016; 12:607-16. [PMID: 26839021 DOI: 10.2217/fon.15.351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The field of prostate cancer has witnessed incredible progress in the last decade, owing to the approval of multiple survival-prolonging treatments for metastatic castration-resistant prostate cancer (mCRPC). Enzalutamide is a nonsteroidal androgen receptor inhibitor that targets multiple steps in the androgen receptor signaling axis. It has been approved for the treatment of mCRPC both in the postdocetaxel and in the chemotherapy-naive settings. We summarize the milestones in the development of enzalutamide in patients with prostate cancer. Special focus is placed on the results of the STRIVE Phase II clinical trial comparing head to head enzalutamide and bicalutamide in patients with nonmetastatic and mCRPC who have failed androgen deprivation and in other ongoing trials in the same setting and in earlier disease phases.
Collapse
Affiliation(s)
- Rosa Nadal
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
| |
Collapse
|
90
|
Sugawara T, Lejeune P, Köhr S, Neuhaus R, Faus H, Gelato KA, Busemann M, Cleve A, Lücking U, von Nussbaum F, Brands M, Mumberg D, Jung K, Stephan C, Haendler B. BAY 1024767 blocks androgen receptor mutants found in castration-resistant prostate cancer patients. Oncotarget 2016; 7:6015-28. [PMID: 26760770 PMCID: PMC4868737 DOI: 10.18632/oncotarget.6864] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/24/2015] [Indexed: 11/25/2022] Open
Abstract
Androgen receptor (AR) mutations arise in patients developing resistance to hormone deprivation therapies. Here we describe BAY 1024767, a thiohydantoin derivative with strong antagonistic activity against nine AR variants with mutations located in the AR ligand-binding domain (LBD), and against wild-type AR. Antagonism was maintained, though reduced, at increased androgen levels. Anti-tumor efficacy was evidenced in vivo in the KuCaP-1 prostate cancer model which bears the W741C bicalutamide resistance mutation and in the syngeneic prostate cancer rat model Dunning R3327-G. The prevalence of six selected AR mutations was determined in plasma DNA originating from 100 resistant patients and found to be at least 12%. Altogether the results show BAY 1024767 to be a strong antagonist for several AR mutants linked to therapy resistance, which opens the door for next-generation compounds that can benefit patients based on their mutation profile.
Collapse
Affiliation(s)
| | | | - Silke Köhr
- Global Drug Discovery, Bayer Pharma AG, Berlin, Germany
| | | | | | | | | | - Arwed Cleve
- Global Drug Discovery, Bayer Pharma AG, Berlin, Germany
| | | | | | | | | | - Klaus Jung
- Berlin Institute of Urologic Research, Berlin, Germany
- Department of Urology, Charité University Hospital, Berlin, Germany
| | - Carsten Stephan
- Berlin Institute of Urologic Research, Berlin, Germany
- Department of Urology, Charité University Hospital, Berlin, Germany
| | | |
Collapse
|
91
|
Chandrasekar T, Yang JC, Gao AC, Evans CP. Mechanisms of resistance in castration-resistant prostate cancer (CRPC). Transl Androl Urol 2016; 4:365-80. [PMID: 26814148 PMCID: PMC4708226 DOI: 10.3978/j.issn.2223-4683.2015.05.02] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite advances in prostate cancer diagnosis and management, morbidity from prostate cancer remains high. Approximately 20% of men present with advanced or metastatic disease, while 29,000 men continue to die of prostate cancer each year. Androgen deprivation therapy (ADT) has been the standard of care for initial management of advanced or metastatic prostate cancer since Huggins and Hodges first introduced the concept of androgen-dependence in 1972, but progression to castration-resistant prostate cancer (CRPC) occurs within 2-3 years of initiation of ADT. CRPC, previously defined as hormone-refractory prostate cancer, is now understood to still be androgen dependent. Multiple mechanisms of resistance help contribute to the progression to castration resistant disease, and the androgen receptor (AR) remains an important driver in this progression. These mechanisms include AR amplification and hypersensitivity, AR mutations leading to promiscuity, mutations in coactivators/corepressors, androgen-independent AR activation, and intratumoral and alternative androgen production. More recently, identification of AR variants (ARVs) has been established as another mechanism of progression to CRPC. Docetaxel chemotherapy has historically been the first-line treatment for CRPC, but in recent years, newer agents have been introduced that target some of these mechanisms of resistance, thereby providing additional survival benefit. These include AR signaling inhibitors such as enzalutamide (Xtandi, ENZA, MDV-3100) and CYP17A1 inhibitors such as abiraterone acetate (Zytiga). Ultimately, these agents will also fail to suppress CRPC. While some of the mechanisms by which these agents fail are unique, many share similarities to the mechanisms contributing to CRPC progression. Understanding these mechanisms of resistance to ADT and currently approved CRPC treatments will help guide future research into targeted therapies.
Collapse
Affiliation(s)
| | - Joy C Yang
- Department of Urology, University of California, Davis, CA, USA
| | - Allen C Gao
- Department of Urology, University of California, Davis, CA, USA
| | | |
Collapse
|
92
|
Zarif JC, Lamb LE, Schulz VV, Nollet EA, Miranti CK. Androgen receptor non-nuclear regulation of prostate cancer cell invasion mediated by Src and matriptase. Oncotarget 2016; 6:6862-76. [PMID: 25730905 PMCID: PMC4466655 DOI: 10.18632/oncotarget.3119] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 01/08/2015] [Indexed: 11/25/2022] Open
Abstract
Castration-resistant prostate cancers still depend on nuclear androgen receptor (AR) function despite their lack of dependence on exogenous androgen. Second generation anti-androgen therapies are more efficient at blocking nuclear AR; however resistant tumors still develop. Recent studies indicate Src is highly active in these resistant tumors. By manipulating AR activity in several different prostate cancer cell lines through RNAi, drug treatment, and the use of a nuclear-deficient AR mutant, we demonstrate that androgen acting on cytoplasmic AR rapidly stimulates Src tyrosine kinase via a non-genomic mechanism. Cytoplasmic AR, acting through Src enhances laminin integrin-dependent invasion. Active Matriptase, which cleaves laminin, is elevated within minutes after androgen stimulation, and is subsequently shed into the medium. Matriptase activation and shedding induced by cytoplasmic AR is dependent on Src. Concomitantly, CDCP1/gp140, a Matriptase and Src substrate that controls integrin-based migration, is activated. However, only inhibition of Matriptase, but not CDCP1, suppresses the AR/Src-dependent increase in invasion. Matriptase, present in conditioned medium from AR-stimulated cells, is sufficient to enhance invasion in the absence of androgen. Thus, invasion is stimulated by a rapid but sustained increase in Src activity, mediated non-genomically by cytoplasmic AR, leading to rapid activation and shedding of the laminin protease Matriptase.
Collapse
Affiliation(s)
- Jelani C Zarif
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA.,Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
| | - Laura E Lamb
- Department of Urology, Beaumont Health System - Research Institute, Royal Oak, MI 48073, USA
| | - Veronique V Schulz
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Eric A Nollet
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA.,Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Cindy K Miranti
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
93
|
Abstract
Great strides have been made in the treatment of castration-resistant prostate cancer (CRPC) with the development of new antiandrogens (enzalutamide) and more potent androgen synthesis inhibitors (abiraterone) that have both improved patient outcomes. These new drugs have also helped unravel the complex biology of androgen-androgen receptor driven prostate cancer and brought into prominence various mechanisms triggering the development of drug resistance and tumour cell survival despite use of androgen deprivation therapy (ADT). The complex role of glucocorticoids in the treatment, management and progression of patients with CRPC is integral to these advances. Historically, glucocorticoid treatment has resulted in both subjective and objective responses in patients with advanced-stage prostate cancer. With the use of these new therapeutic agents, however, unexpected glucocorticoid-related mechanisms that can cause iatrogenic stimulation of prostate cancer growth have emerged, which might contribute to drug resistance and disease progression despite optimal ADT. For example, the upregulation of glucocorticoid receptors (GRs) during enzalutamide therapy results in glucocorticoid-GR-mediated regulation of androgen target genes, leading to escape from enzalutamide blockade. Thus, understanding the biological role of glucocorticoids in patients with prostate cancer is of major importance in the era of new and evolving antiandrogen therapies.
Collapse
|
94
|
Chandrasekar T, Yang JC, Gao AC, Evans CP. Targeting molecular resistance in castration-resistant prostate cancer. BMC Med 2015; 13:206. [PMID: 26329698 PMCID: PMC4556222 DOI: 10.1186/s12916-015-0457-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 08/19/2015] [Indexed: 12/20/2022] Open
Abstract
Multiple mechanisms of resistance contribute to the inevitable progression of hormone-sensitive prostate cancer to castration-resistant prostate cancer (CRPC). Currently approved therapies for CRPC include systemic chemotherapy (docetaxel and cabazitaxel) and agents targeting the resistance pathways leading to CRPC, including enzalutamide and abiraterone. While there is significant survival benefit, primary and secondary resistance to these therapies develops rapidly. Up to one-third of patients have primary resistance to enzalutamide and abiraterone; the remaining patients eventually progress on treatment. Understanding the mechanisms of resistance resulting in progression as well as identifying new targetable pathways remains the focus of current prostate cancer research. We review current knowledge of mechanisms of resistance to the currently approved treatments, development of adjunctive therapies, and identification of new pathways being targeted for therapeutic purposes.
Collapse
Affiliation(s)
| | - Joy C Yang
- Department of Urology, University of California, Davis, USA.
| | - Allen C Gao
- Department of Urology, University of California, Davis, USA.
| | - Christopher P Evans
- Department of Urology, University of California, Davis, USA. .,, 4860 Y Street, Suite 3500, Sacramento, CA, 95817, USA.
| |
Collapse
|
95
|
Pihlajamaa P, Sahu B, Jänne OA. Determinants of Receptor- and Tissue-Specific Actions in Androgen Signaling. Endocr Rev 2015; 36:357-84. [PMID: 26052734 DOI: 10.1210/er.2015-1034] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The physiological androgens testosterone and 5α-dihydrotestosterone regulate the development and maintenance of primary and secondary male sexual characteristics through binding to the androgen receptor (AR), a ligand-dependent transcription factor. In addition, a number of nonreproductive tissues of both genders are subject to androgen regulation. AR is also a central target in the treatment of prostate cancer. A large number of studies over the last decade have characterized many regulatory aspects of the AR pathway, such as androgen-dependent transcription programs, AR cistromes, and coregulatory proteins, mostly in cultured cells of prostate cancer origin. Moreover, recent work has revealed the presence of pioneer/licensing factors and chromatin modifications that are important to guide receptor recruitment onto appropriate chromatin loci in cell lines and in tissues under physiological conditions. Despite these advances, current knowledge related to the mechanisms responsible for receptor- and tissue-specific actions of androgens is still relatively limited. Here, we review topics that pertain to these specificity issues at different levels, both in cultured cells and tissues in vivo, with a particular emphasis on the nature of the steroid, the response element sequence, the AR cistromes, pioneer/licensing factors, and coregulatory proteins. We conclude that liganded AR and its DNA-response elements are required but are not sufficient for establishment of tissue-specific transcription programs in vivo, and that AR-selective actions over other steroid receptors rely on relaxed rather than increased stringency of cis-elements on chromatin.
Collapse
Affiliation(s)
- Päivi Pihlajamaa
- Department of Physiology (P.P., B.S., O.A.J.), and Research Programs Unit, Genome-Scale Biology (P.P., B.S.), Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| | - Biswajyoti Sahu
- Department of Physiology (P.P., B.S., O.A.J.), and Research Programs Unit, Genome-Scale Biology (P.P., B.S.), Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| | - Olli A Jänne
- Department of Physiology (P.P., B.S., O.A.J.), and Research Programs Unit, Genome-Scale Biology (P.P., B.S.), Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
96
|
Modena A, Ciccarese C, Fantinel E, Bimbatti D, Tortora G, Massari F. Metastatic castration-resistant prostate cancer: targeting the mechanisms of resistance to abiraterone acetate and enzalutamide. Expert Rev Anticancer Ther 2015; 15:1037-48. [DOI: 10.1586/14737140.2015.1063423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
97
|
Rodriguez-Vida A, Galazi M, Rudman S, Chowdhury S, Sternberg CN. Enzalutamide for the treatment of metastatic castration-resistant prostate cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3325-39. [PMID: 26170619 PMCID: PMC4492664 DOI: 10.2147/dddt.s69433] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In recent years, several nonhormonal and hormonal agents, including enzalutamide, have been approved for the treatment of metastatic castration-resistant prostate cancer (CRPC) on the basis of improved overall survival in prospective clinical trials. The incorporation of these agents has revolutionized the treatment of CRPC but has also raised the question of what is the ideal sequence of administering them. Enzalutamide is a nonsteroidal second-generation antiandrogen that has been approved for the treatment of metastatic CRPC both in the post-docetaxel and chemotherapy-naïve settings. This article reviews the pharmacological characteristics of enzalutamide, the efficacy studies which led to its approval, its safety profile, and quality of life-related parameters as well as its place in the sequential treatment and management of metastatic prostate cancer.
Collapse
Affiliation(s)
- Alejo Rodriguez-Vida
- Medical Oncology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Myria Galazi
- Medical Oncology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Sarah Rudman
- Medical Oncology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Simon Chowdhury
- Medical Oncology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Cora N Sternberg
- Medical Oncology Department, San Camillo and Forlanini Hospitals, Rome, Italy
| |
Collapse
|
98
|
Hsieh CL, Botta G, Gao S, Li T, Van Allen EM, Treacy DJ, Cai C, He HH, Sweeney CJ, Brown M, Balk SP, Nelson PS, Garraway LA, Kantoff PW. PLZF, a tumor suppressor genetically lost in metastatic castration-resistant prostate cancer, is a mediator of resistance to androgen deprivation therapy. Cancer Res 2015; 75:1944-8. [PMID: 25808865 DOI: 10.1158/0008-5472.can-14-3602] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/16/2015] [Indexed: 01/18/2023]
Abstract
Whole-exome sequencing of metastatic castration-resistant prostate cancer (mCRPC) reveals that 5% to 7% of tumors harbor promyelocytic leukemia zinc finger (PLZF) protein homozygous deletions. PLZF is a canonical androgen-regulated putative tumor suppressor gene whose expression is inhibited by androgen deprivation therapy (ADT). Here, we demonstrate that knockdown of PLZF expression promotes a CRPC and enzalutamide-resistant phenotype in prostate cancer cells. Reintroduction of PLZF expression is sufficient to reverse androgen-independent growth mediated by PLZF depletion. PLZF loss enhances CRPC tumor growth in a xenograft model. Bioinformatic analysis of the PLZF cistrome shows that PLZF negatively regulates multiple pathways, including the MAPK pathway. Accordingly, our data support an oncogenic program activated by ADT. This acquired mechanism together with the finding of genetic loss in CRPC implicates PLZF inactivation as a mechanism promoting ADT resistance and the CRPC phenotype.
Collapse
Affiliation(s)
| | - Ginevra Botta
- Department of Medical Oncology, and Broad Institute, Cambridge, Massachusetts
| | - Shuai Gao
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | | | - Eliezer M Van Allen
- Department of Medical Oncology, and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | | | - Changmeng Cai
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Housheng Hansen He
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Myles Brown
- Department of Medical Oncology, and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Steven P Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Peter S Nelson
- Division of Clinical Research, Fred Hutchinson Cancer Research Center and Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Levi A Garraway
- Department of Medical Oncology, and Broad Institute, Cambridge, Massachusetts.
| | | |
Collapse
|
99
|
Locke JA, Dal Pra A, Supiot S, Warde P, Bristow RG. Synergistic action of image-guided radiotherapy and androgen deprivation therapy. Nat Rev Urol 2015; 12:193-204. [PMID: 25800395 DOI: 10.1038/nrurol.2015.50] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The combined use of androgen deprivation therapy (ADT) and image-guided radiotherapy (IGRT) can improve overall survival in aggressive, localized prostate cancer. However, owing to the adverse effects of prolonged ADT, it is imperative to identify the patients who would benefit from this combined-modality therapy relative to the use of IGRT alone. Opportunities exist for more personalized approaches in treating aggressive, locally advanced prostate cancer. Biomarkers--such as disseminated tumour cells, circulating tumour cells, genomic signatures and molecular imaging techniques--could identify the patients who are at greatest risk for systemic metastases and who would benefit from the addition of systemic ADT. By contrast, when biomarkers of systemic disease are not present, treatment could proceed using local IGRT alone. The choice of drug, treatment duration and timing of ADT relative to IGRT could be predicated on these personalized approaches to prostate cancer medicine. These novel treatment intensification and reduction strategies could result in improved prostate-cancer-specific survival and overall survival, without incurring the added expense of metabolic syndrome and other adverse effects of ADT in all patients.
Collapse
Affiliation(s)
- Jennifer A Locke
- Department of Urologic Sciences, University of British Columbia, Gordon &Leslie Diamond Health Care Centre, Level 6, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada
| | - Alan Dal Pra
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 4, CH-3010 Bern, Switzerland
| | - Stéphane Supiot
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes-St-Herblain, 8 quai Moncousu, BP 70721, 44000 Nantes, France
| | - Padraig Warde
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - Robert G Bristow
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
100
|
Wang JQ, DeChalus A, Chatterjee DN, Keller ET, Mizokami A, Camussi G, Mendelsohn AR, Renzulli JF, Quesenberry PJ, Chatterjee D. Extracellular Vesicle-Mediated Reversal of Paclitaxel Resistance in Prostate Cancer. Crit Rev Oncog 2015; 20:407-17. [PMID: 27279238 PMCID: PMC5331882 DOI: 10.1615/critrevoncog.v20.i5-6.120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Prostate cancer (PCa) is the most common solid tumor in males and the second leading cause of cancer-related deaths in males in the United States. The current first line therapy for metastatic PCa is androgen deprivation therapy and is initially effective against the disease. However, castrate resistant prostate cancer (CRPC) develops in many men within 18-36 months, rendering this treatment ineffective. Chemotherapy, with a class of drugs known as taxanes is the standard-of-care cytotoxic option in metastatic castrate resistant PCa (mCRPC). However, the overall survival advantage for chemotherapy in mCRPC is only 2.2 months and the cancer cells often become resistant to these drugs as well. Once patients fail chemotherapy the progression to death is inevitable. Extracellular vesicles (EVs) are involved in cell signaling and play a role in cancer progression. Previous work has demonstrated that EVs are involved in the development of drug resistance in cancer cells. We report the reversal of taxane resistance and tumorigenic phenotype in PCa cells after EVs treatment. This study suggests that EVs represent a potentially novel therapeutic treatment option for CRPC.
Collapse
Affiliation(s)
- Justin Q. Wang
- Department of Medicine, Rhode Island Hospital, Providence, RI
| | - Austin DeChalus
- Department of Medicine, Rhode Island Hospital, Providence, RI
| | | | - Evan T. Keller
- Department of Urology, University of Michigan, Ann Arbor, MI
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI
| | | | - Giovanni Camussi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | | | - Joseph F. Renzulli
- Department of Urology, Miriam Hospital, The Alpert Medical School of Brown University, Providence, RI
| | | | | |
Collapse
|