1
|
Adzavon YM, Culig Z, Sun Z. Interactions between androgen and IGF1 axes in prostate tumorigenesis. Nat Rev Urol 2025; 22:268-275. [PMID: 39375467 DOI: 10.1038/s41585-024-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/09/2024]
Abstract
Androgen signalling through the androgen receptor (AR) is essential for prostate tumorigenesis. However, androgen signalling pathways also interact with other growth factor-mediated signalling pathways to regulate the prostatic cell cycle, differentiation, apoptosis and proliferation in the initiation and progression of prostate cancer. Insulin-like growth factor 1 (IGF1) is one of the most prominent growth factors in prostate tumorigenesis. Clinical and experimental evidence has demonstrated that IGF1 signalling supports both androgen-dependent and androgen-independent prostate tumorigenesis, suggesting that improved understanding of the interactions between the IGF1 and androgen axes might aid the development of new therapeutic strategies. Available data have shown a dynamic role of androgen-AR signalling in the activation of IGF1-signalling pathways by augmenting transcription of the IGF1 receptor in prostatic basal epithelial cells and by increasing IGF1 secretion through the suppression of IGF-binding protein 3 expression in prostatic stromal cells. In turn, IGF1 stimulates Wnt-β-catenin signalling in prostatic basal progenitors to promote prostatic oncogenic transformation and prostate cancer development. These findings highlight the cooperative, autocrine and paracrine interactions that underlie the oncogenic effects of androgens and IGF1 and open up new opportunities for therapeutic targeting.
Collapse
Affiliation(s)
- Yao Mawulikplimi Adzavon
- Department of Cell Biology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Oncology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zoran Culig
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zijie Sun
- Department of Cell Biology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
2
|
Han D, Labaf M, Zhao Y, Owiredu J, Zhang S, Patel K, Venkataramani K, Steinfeld JS, Han W, Li M, Liu M, Wang Z, Besschetnova A, Patalano S, Mulhearn MJ, Macoska JA, Yuan X, Balk SP, Nelson PS, Plymate SR, Gao S, Siegfried KR, Liu R, Stangis MM, Foxa G, Czernik PJ, Williams BO, Zarringhalam K, Li X, Cai C. Androgen receptor splice variants drive castration-resistant prostate cancer metastasis by activating distinct transcriptional programs. J Clin Invest 2024; 134:e168649. [PMID: 38687617 PMCID: PMC11142739 DOI: 10.1172/jci168649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
One critical mechanism through which prostate cancer (PCa) adapts to treatments targeting androgen receptor (AR) signaling is the emergence of ligand-binding domain-truncated and constitutively active AR splice variants, particularly AR-V7. While AR-V7 has been intensively studied, its ability to activate distinct biological functions compared with the full-length AR (AR-FL), and its role in regulating the metastatic progression of castration-resistant PCa (CRPC), remain unclear. Our study found that, under castrated conditions, AR-V7 strongly induced osteoblastic bone lesions, a response not observed with AR-FL overexpression. Through combined ChIP-seq, ATAC-seq, and RNA-seq analyses, we demonstrated that AR-V7 uniquely accesses the androgen-responsive elements in compact chromatin regions, activating a distinct transcription program. This program was highly enriched for genes involved in epithelial-mesenchymal transition and metastasis. Notably, we discovered that SOX9, a critical metastasis driver gene, was a direct target and downstream effector of AR-V7. Its protein expression was dramatically upregulated in AR-V7-induced bone lesions. Moreover, we found that Ser81 phosphorylation enhanced AR-V7's pro-metastasis function by selectively altering its specific transcription program. Blocking this phosphorylation with CDK9 inhibitors impaired the AR-V7-mediated metastasis program. Overall, our study has provided molecular insights into the role of AR splice variants in driving the metastatic progression of CRPC.
Collapse
Affiliation(s)
- Dong Han
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Maryam Labaf
- Center for Personalized Cancer Therapy
- Department of Mathematics, University of Massachusetts Boston, Boston, Massachusetts, USA
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yawei Zhao
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Jude Owiredu
- Department of Cell & Developmental Biology, Weill Cornell Medical College, New York, New York, USA
| | - Songqi Zhang
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Krishna Patel
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | | | | | - Wanting Han
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Muqing Li
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Mingyu Liu
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Zifeng Wang
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | | | - Susan Patalano
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | | | - Jill A. Macoska
- Center for Personalized Cancer Therapy
- Department of Biology, and
| | - Xin Yuan
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven P. Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter S. Nelson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stephen R. Plymate
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Veterans Affairs Puget Sound Health Care System, Geriatric Research and Education Clinical Center (VAPSHCS-GRECC), Seattle, Washington, USA
| | - Shuai Gao
- Department of Cell Biology and Anatomy and
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| | | | - Ruihua Liu
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Mary M. Stangis
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Gabrielle Foxa
- Department of Cell Biology, and Core Technologies and Services, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Piotr J. Czernik
- Department of Orthopaedic Surgery, MicroCT and Skeletal Research Core Facility, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Bart O. Williams
- Department of Cell Biology, and Core Technologies and Services, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Kourosh Zarringhalam
- Center for Personalized Cancer Therapy
- Department of Mathematics, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Xiaohong Li
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, USA
| | - Changmeng Cai
- Center for Personalized Cancer Therapy
- Department of Biology, and
| |
Collapse
|
3
|
Zhang C, Ren J, Kang Y, Chang D. Case report and literature review of rezvilutamide in the treatment of hormone-sensitive prostate cancer. Front Oncol 2024; 14:1374039. [PMID: 38577344 PMCID: PMC10991726 DOI: 10.3389/fonc.2024.1374039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/04/2024] [Indexed: 04/06/2024] Open
Abstract
Background Prostate cancer represents a major health concern worldwide, with the treatment of metastatic hormone-sensitive prostate cancer (mHSPC) and locally advanced prostate cancer posing a particular challenge. Rezvilutamide, a new androgen receptor antagonist from China, has shown early promise; however, its real-world effectiveness and safety profile require further evidence. This case series evaluates the preliminary clinical outcomes of rezvilutamide in combination with androgen deprivation therapy (ADT), focusing on PSA response and radiological findings across various stages of prostate cancer in four patients. Case description Case 1 details a 68-year-old male with low-volume mHSPC who exhibited a positive therapeutic response, demonstrated by decreasing PSA levels and improved radiographic results, despite experiencing mild side effects related to the drug. Case 2 describes a 71-year-old male with high-volume mHSPC who had a favorable outcome, with no significant changes in tumor size or metastatic spread and no negative reactions to the drug. Case 3 involves a 55-year-old male with locally advanced prostate cancer, who saw a reduction in PSA levels and a small decrease in tumor volume, yet with ongoing bladder involvement. Genetic testing showed no significant mutations. Case 4 presents a 74-year-old male with extensive metastatic disease who initially responded to the treatment but later exhibited disease advancement and an ATM gene mutation, signaling a shift to metastatic castration-resistant prostate cancer (mCRPC). This finding underscores the crucial role of genetic testing in directing future treatment, with therapies such as olaparib or chemotherapy being advised. Conclusions Rezvilutamide has shown a potential benefit in the management of mHSPC and locally advanced prostate cancer, generally with a mild safety profile. Initial positive responses, particularly in PSA decline and radiographic progression, are promising. Nevertheless, the varying responses, notably concerning genetic mutations, highlight the necessity for tailored treatment approaches. Due to the small cohort and brief follow-up period, more extensive research with larger populations and prolonged monitoring is essential to conclusively determine the benefits and safety of rezvilutamide. The utilization of genetic insights is key to refining treatment decisions and enhancing outcomes for patients with advanced prostate cancer.
Collapse
Affiliation(s)
| | | | | | - Dehui Chang
- Department of Urology, The 940 Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, Gansu, China
| |
Collapse
|
4
|
Katleba KD, Ghosh PM, Mudryj M. Beyond Prostate Cancer: An Androgen Receptor Splice Variant Expression in Multiple Malignancies, Non-Cancer Pathologies, and Development. Biomedicines 2023; 11:2215. [PMID: 37626712 PMCID: PMC10452427 DOI: 10.3390/biomedicines11082215] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Multiple studies have demonstrated the importance of androgen receptor (AR) splice variants (SVs) in the progression of prostate cancer to the castration-resistant phenotype and their utility as a diagnostic. However, studies on AR expression in non-prostatic malignancies uncovered that AR-SVs are expressed in glioblastoma, breast, salivary, bladder, kidney, and liver cancers, where they have diverse roles in tumorigenesis. AR-SVs also have roles in non-cancer pathologies. In granulosa cells from women with polycystic ovarian syndrome, unique AR-SVs lead to an increase in androgen production. In patients with nonobstructive azoospermia, testicular Sertoli cells exhibit differential expression of AR-SVs, which is associated with impaired spermatogenesis. Moreover, AR-SVs have been identified in normal cells, including blood mononuclear cells, neuronal lipid rafts, and the placenta. The detection and characterization of AR-SVs in mammalian and non-mammalian species argue that AR-SV expression is evolutionarily conserved and that AR-SV-dependent signaling is a fundamental regulatory feature in multiple cellular contexts. These discoveries argue that alternative splicing of the AR transcript is a commonly used mechanism that leads to an expansion in the repertoire of signaling molecules needed in certain tissues. Various malignancies appropriate this mechanism of alternative AR splicing to acquire a proliferative and survival advantage.
Collapse
Affiliation(s)
- Kimberley D. Katleba
- Veterans Affairs-Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA; (K.D.K.); (P.M.G.)
- Department of Medical Microbiology and Immunology, 1 Shields Avenue, UC Davis, Davis, CA 95616, USA
| | - Paramita M. Ghosh
- Veterans Affairs-Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA; (K.D.K.); (P.M.G.)
- Department of Urologic Surgery, 4860 Y Street, UC Davis, Sacramento, CA 95718, USA
- Department of Biochemistry and Molecular Medicine, 1 Shields Avenue, UC Davis, Davis, CA 95616, USA
| | - Maria Mudryj
- Veterans Affairs-Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA; (K.D.K.); (P.M.G.)
- Department of Medical Microbiology and Immunology, 1 Shields Avenue, UC Davis, Davis, CA 95616, USA
| |
Collapse
|
5
|
Biernacka KM, Barker R, Sewell A, Bahl A, Perks CM. A role for androgen receptor variant 7 in sensitivity to therapy: Involvement of IGFBP-2 and FOXA1. Transl Oncol 2023; 34:101698. [PMID: 37307644 PMCID: PMC10276180 DOI: 10.1016/j.tranon.2023.101698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/10/2023] [Accepted: 05/21/2023] [Indexed: 06/14/2023] Open
Abstract
Prostate cancer (PCa) is one of the leading causes of cancer-related deaths in men. Localised PCa can be treated effectively, but most patients relapse/progress to more aggressive disease. One possible mechanism underlying this progression is alternative splicing of the androgen receptor, with AR variant 7(ARV7) considered to play a major role. Using viability assays, we confirmed that ARV7-positive PCa cells were less sensitive to treatment with cabazitaxel and an anti-androgen-enzalutamide. Also, using live-holographic imaging, we showed that PCa cells with ARV7 exhibited an increased rate of cell division, proliferation, and motility, which could potentially contribute to a more aggressive phenotype. Furthermore, protein analysis demonstrated that ARV7 knock-down was associated with a decrease in insulin-like growth factor-2 (IGFBP-2) and forkhead box protein A1(FOXA1). This correlation was confirmed in-vivo using PCa tissue samples. Spearman rank correlation analysis showed significant positive associations between ARV7 and IGFBP-2 or FOXA1 in tissue from patients with PCa. This association was not present with the AR. These data suggest an interplay of FOXA1 and IGFBP-2 with ARV7-mediated acquisition of an aggressive prostate cancer phenotype.
Collapse
Affiliation(s)
- K M Biernacka
- Cancer Endocrinology Group, Translational Health Sciences, University of Bristol Southmead Hospital, BS10 5NB, Bristol, UK
| | - R Barker
- Cancer Endocrinology Group, Translational Health Sciences, University of Bristol Southmead Hospital, BS10 5NB, Bristol, UK
| | - A Sewell
- Department of Cellular Pathology, North Bristol NHS Trust, Southmead Hospital, Bristol, UK
| | - A Bahl
- Bristol Haematology and Oncology Centre, Department of Clinical Oncology, University Hospitals Bristol, Bristol BS2 8ED, UK
| | - C M Perks
- Cancer Endocrinology Group, Translational Health Sciences, University of Bristol Southmead Hospital, BS10 5NB, Bristol, UK.
| |
Collapse
|
6
|
Androgen receptor variant 7 exacerbates hepatocarcinogenesis in a c-MYC-driven mouse HCC model. Oncogenesis 2023; 12:4. [PMID: 36746917 PMCID: PMC9902460 DOI: 10.1038/s41389-023-00449-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 02/08/2023] Open
Abstract
Androgen receptor variant 7 (AR-V7), an AR isoform with a truncated ligand-binding domain, functions as a transcription factor in an androgen-independent manner. AR-V7 is expressed in a subpopulation of hepatocellular carcinoma (HCC), however, its role(s) in this cancer is undefined. In this study, we investigated the potential roles of AR-V7 in hepatocarcinogenesis in vivo in a c-MYC-driven mouse HCC model generated by the hydrodynamic tail-vein injection system. The impacts of AR-V7 on gene expression in mouse HCC were elucidated by RNA-seq transcriptome and ontology analyses. The results showed that AR-V7 significantly exacerbated the c-MYC-mediated oncogenesis in the livers of both sexes. The transcriptome and bioinformatics analyses revealed that AR-V7 and c-MYC synergistically altered the gene sets involved in various cancer-related biological processes, particularly in lipid and steroid/sterol metabolisms. Importantly, AR-V7 suppressed a tumor suppressor Claudin 7 expression, upregulated by c-MYC overexpression via the p53 signaling pathway. Claudin 7 overexpression significantly suppressed the c-MYC-driven HCC development under p53-deficient conditions. Our results suggest that the AR-V7 exacerbates the c-MYC-driven hepatocarcinogenesis by potentiating the oncogenic roles and minimizing the anti-oncogenic functions of c-MYC. Since AR-V7 is expressed in a subpopulation of HCC cases, it could contribute to the inter- and intra-heterogeneity of HCC.
Collapse
|
7
|
Kim WK, Olson AW, Mi J, Wang J, Lee DH, Le V, Hiroto A, Aldahl J, Nenninger CH, Buckley AJ, Cardiff R, You S, Sun Z. Aberrant androgen action in prostatic progenitor cells induces oncogenesis and tumor development through IGF1 and Wnt axes. Nat Commun 2022; 13:4364. [PMID: 35902588 PMCID: PMC9334353 DOI: 10.1038/s41467-022-32119-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 07/18/2022] [Indexed: 12/26/2022] Open
Abstract
Androgen/androgen receptor (AR) signaling pathways are essential for prostate tumorigenesis. However, the fundamental mechanisms underlying the AR functioning as a tumor promoter in inducing prostatic oncogenesis still remain elusive. Here, we demonstrate that a subpopulation of prostatic Osr1 (odd skipped-related 1)-lineage cells functions as tumor progenitors in prostate tumorigenesis. Single cell transcriptomic analyses reveal that aberrant AR activation in these cells elevates insulin-like growth factor 1 (IGF1) signaling pathways and initiates oncogenic transformation. Elevating IGF1 signaling further cumulates Wnt/β-catenin pathways in transformed cells to promote prostate tumor development. Correlations between altered androgen, IGF1, and Wnt/β-catenin signaling are also identified in human prostate cancer samples, uncovering a dynamic regulatory loop initiated by the AR through prostate cancer development. Co-inhibition of androgen and Wnt-signaling pathways significantly represses the growth of AR-positive tumor cells in both ex-vivo and in-vivo, implicating co-targeting therapeutic strategies for these pathways to treat advanced prostate cancer.
Collapse
Affiliation(s)
- Won Kyung Kim
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Adam W Olson
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jiaqi Mi
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jinhui Wang
- Integrative Genomics Core, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Dong-Hoon Lee
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Vien Le
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Alex Hiroto
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Joseph Aldahl
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Christian H Nenninger
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Alyssa J Buckley
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Robert Cardiff
- Center for Comparative Medicine, University of California at Davis, Davis, CA, USA
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zijie Sun
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
8
|
Huang J, Lin B, Li B. Anti-Androgen Receptor Therapies in Prostate Cancer: A Brief Update and Perspective. Front Oncol 2022; 12:865350. [PMID: 35372068 PMCID: PMC8965587 DOI: 10.3389/fonc.2022.865350] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/17/2022] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer is a major health issue in western countries and is the second leading cause of cancer death in American men. Prostate cancer depends on the androgen receptor (AR), a transcriptional factor critical for prostate cancer growth and progression. Castration by surgery or medical treatment reduces androgen levels, resulting in prostatic atrophy and prostate cancer regression. Thus, metastatic prostate cancers are initially managed with androgen deprivation therapy. Unfortunately, prostate cancers rapidly relapse after castration therapy and progress to a disease stage called castration-resistant prostate cancer (CRPC). Currently, clinical treatment for CRPCs is focused on suppressing AR activity with antagonists like Enzalutamide or by reducing androgen production with Abiraterone. In clinical practice, these treatments fail to yield a curative benefit in CRPC patients in part due to AR gene mutations or splicing variations, resulting in AR reactivation. It is conceivable that eliminating the AR protein in prostate cancer cells is a promising solution to provide a potential curative outcome. Multiple strategies have emerged, and several potent agents that reduce AR protein levels were reported to eliminate xenograft tumor growth in preclinical models via distinct mechanisms, including proteasome-mediated degradation, heat-shock protein inhibition, AR splicing suppression, blockage of AR nuclear localization, AR N-terminal suppression. A few small chemical compounds are undergoing clinical trials combined with existing AR antagonists. AR protein elimination by enhanced protein or mRNA degradation is a realistic solution for avoiding AR reactivation during androgen deprivation therapy in prostate cancers.
Collapse
Affiliation(s)
- Jian Huang
- Pathological Diagnosis and Research Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Biyun Lin
- Pathological Diagnosis and Research Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
9
|
Öther-Gee Pohl S, Myant KB. Alternative RNA splicing in tumour heterogeneity, plasticity and therapy. Dis Model Mech 2022; 15:dmm049233. [PMID: 35014671 PMCID: PMC8764416 DOI: 10.1242/dmm.049233] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alternative splicing is a process by which a single gene is able to encode multiple different protein isoforms. It is regulated by the inclusion or exclusion of introns and exons that are joined in different patterns prior to protein translation, thus enabling transcriptomic and proteomic diversity. It is now widely accepted that alternative splicing is dysregulated across nearly all cancer types. This widespread dysregulation means that nearly all cellular processes are affected - these include processes synonymous with the hallmarks of cancer - evasion of apoptosis, tissue invasion and metastasis, altered cellular metabolism, genome instability and drug resistance. Emerging evidence indicates that the dysregulation of alternative splicing also promotes a permissive environment for increased tumour heterogeneity and cellular plasticity. These are fundamental regulators of a patient's response to therapy. In this Review, we introduce the mechanisms of alternative splicing and the role of aberrant splicing in cancer, with particular focus on newfound evidence of alternative splicing promoting tumour heterogeneity, cellular plasticity and altered metabolism. We discuss recent in vivo models generated to study alternative splicing and the importance of these for understanding complex tumourigenic processes. Finally, we review the effects of alternative splicing on immune evasion, cell death and genome instability, and how targeting these might enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | - Kevin B. Myant
- Cancer Research UK Edinburgh Centre, Institute of Genetics of Cancer, The University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| |
Collapse
|
10
|
Liu Y, Liu X, Lin C, Jia X, Zhu H, Song J, Zhang Y. Noncoding RNAs regulate alternative splicing in Cancer. J Exp Clin Cancer Res 2021; 40:11. [PMID: 33407694 PMCID: PMC7789004 DOI: 10.1186/s13046-020-01798-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
AS (alternative splicing) is a fundamental process by which a gene can generate multiple distinct mRNA transcripts to increase protein diversity. Defects in AS influence the occurrence and development of many diseases, including cancers, and are frequently found to participate in various aspects of cancer biology, such as promoting invasion, metastasis, apoptosis resistance and drug resistance. NcRNAs (noncoding RNAs) are an abundant class of RNAs that do not encode proteins. NcRNAs include miRNAs (microRNAs), lncRNAs (long noncoding RNAs), circRNAs (circular RNAs) and snRNAs (small nuclear RNAs) and have been proven to act as regulatory molecules that mediate cancer processes through AS. NcRNAs can directly or indirectly influence a plethora of molecular targets to regulate cis-acting elements, trans-acting factors, or pre-mRNA transcription at multiple levels, affecting the AS process and generating alternatively spliced isoforms. Consequently, ncRNA-mediated AS outcomes affect multiple cellular signaling pathways that promote or suppress cancer progression. In this review, we summarize the current mechanisms by which ncRNAs regulate AS in cancers and discuss their potential clinical applications as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yunze Liu
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
- Department of Traditional Chinese Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Xin Liu
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Changwei Lin
- Department of Gastrointestinal Surgery, the Third XiangYa Hospital of Central South University, Changsha, 410013, China
| | - Xianhong Jia
- Department of Traditional Chinese Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Hongmei Zhu
- Department of Traditional Chinese Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Jun Song
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.
| | - Yi Zhang
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.
| |
Collapse
|
11
|
Lv Y, Zhang X, Chen L. Suspension state regulates epithelial-to-mesenchymal transition and stemness of breast tumor cells. Biotechnol Lett 2021; 43:561-578. [PMID: 33386502 DOI: 10.1007/s10529-020-03074-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The mechanical forces on circulating tumor cells (CTCs) should not be ignored in blood and it is more essential that CTCs can overcome and utilize the mechanical interaction to acquire the ability of distant metastasis. At present there are few studies on how suspension mechanics regulates the behavior of tumor cells. The aim of the study was to explore the effects of suspension state on the epithelial-mesenchymal transition (EMT) and stemness of breast CTCs and the molecular mechanisms involved. RESULTS Suspension state could regulate the program of EMT in breast cancer cells, which supported the complex dynamic concept of EMT. It is that the Ras homolog family member A (RhoA)/Rho-associated coiled-coil containing protein kinase 1 (ROCK1) signaling pathway was activated by suspension state in MCF-7 cells instead of MDA-MB-231 cells. In addition, suspension state increased the stemness of breast cancer cells from different aspects. CONCLUSION The study highlighted the emergence of hybrid epithelial/mesenchymal (E/M) state during hematogenous metastasis and the plasticity of CTCs caused by cancer stem cells, further providing novel insights into clinical monitoring of CTCs and therapeutic strategies.
Collapse
Affiliation(s)
- Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, People's Republic of China.
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.
| | - Xiaomei Zhang
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, People's Republic of China
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Lini Chen
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, People's Republic of China
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| |
Collapse
|
12
|
Chen Y, Lan T. Molecular Origin, Expression Regulation, and Biological Function of Androgen Receptor Splicing Variant 7 in Prostate Cancer. Urol Int 2020; 105:337-353. [PMID: 32957106 DOI: 10.1159/000510124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/07/2020] [Indexed: 11/19/2022]
Abstract
The problem of resistance to therapy in prostate cancer (PCa) is multifaceted. Key determinants of drug resistance include tumor burden and growth kinetics, tumor heterogeneity, physical barriers, immune system and microenvironment, undruggable cancer drivers, and consequences of therapeutic pressures. With regard to the fundamental importance of the androgen receptor (AR) in all stages of PCa from tumorigenesis to progression, AR is postulated to have a continued critical role in castration-resistant prostate cancer (CRPC). Suppression of AR signaling mediated by the full-length AR (AR-FL) is the therapeutic goal of all AR-directed therapies. However, AR-targeting agents ultimately lead to AR aberrations that promote PCa progression and drug resistance. Among these AR aberrations, androgen receptor variant 7 (AR-V7) is gaining attention as a potential predictive marker for as well as one of the resistance mechanisms to the most current anti-AR therapies in CRPC. Meanwhile, development of next-generation drugs that directly or indirectly target AR-V7 signaling is urgently needed. In the present review of the current literature, we have summarized the origin, alternative splicing, expression induction, protein conformation, interaction with coregulators, relationship with AR-FL, transcriptional activity, and biological function of AR-V7 in PCa development and therapeutic resistance. We hope this review will help further understand the molecular origin, expression regulation, and role of AR-V7 in the progression of PCa and provide insight into the design of novel selective inhibitors of AR-V7 in PCa treatment.
Collapse
Affiliation(s)
- Ye Chen
- Department of Surgery and Anesthesiology, Joint Logistic Support 940 Hospital of CPLA, Lanzhou, China
| | - Tian Lan
- Department of Urology, Joint Logistic Support 940 Hospital of CPLA, Lanzhou, China,
| |
Collapse
|
13
|
Protein Expression Analysis of an In Vitro Murine Model of Prostate Cancer Progression: Towards Identification of High-Potential Therapeutic Targets. J Pers Med 2020; 10:jpm10030083. [PMID: 32784957 PMCID: PMC7565308 DOI: 10.3390/jpm10030083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Prostate cancer (PC) is the most frequently diagnosed cancer among men worldwide. The poor prognosis of PC is largely due to late diagnosis of the disease when it has progressed to advanced stages marked by androgen-independence. We interrogated proteomic signatures that embody the transition of PC from an androgen-dependent (AD) to an androgen-independent (AI) state. Methods: We have previously established AD and AI murine PC cell lines, PLum-AD and PLum-AI, respectively, which recapitulate primary and progressive PC at phenotypic and subcellular levels. We statistically surveyed global protein expression profiles in these cell lines. Differential profiles were functionally interrogated by pathways and protein–protein interaction network analyses. Results: Protein expression pattern analysis revealed a total of 683 proteins, among which 99 were significantly differentially altered in PLum-AI cells as compared to PLum-AD cells (45 increased and 54 decreased). Principal component analysis (PCA) revealed that the two different cell lines clearly separated apart, indicating a significant proteome expression difference between them. Four of the proteins (vimentin, catalase, EpCAM, and caspase 3) that were differentially expressed in PLum-AI cells compared to PLum-AD cells were subjected to biochemical validation by Western blotting. Biological process gene ontology (GO) analysis of the differentially expressed proteins demonstrated enrichment of biological functions and pathways in PLum-AI cells that are central to PI3 kinase and androgen receptor pathways. Besides, other relevant biological processes that are enriched in PLum-AI cells included cell adhesion and cell migration processes, cell and DNA damage, apoptosis, and cell cycle regulation. Conclusions: Our protein expression analysis of a murine in vitro model of PC progression identified differential protein spots that denote this progression and that comprise high-potential targets for early treatment of PC with a personalized patient-specific approach. Efforts are underway to functionally assess the potential roles of these proteins as therapeutic targets for PC progression.
Collapse
|
14
|
Carceles-Cordon M, Kelly WK, Gomella L, Knudsen KE, Rodriguez-Bravo V, Domingo-Domenech J. Cellular rewiring in lethal prostate cancer: the architect of drug resistance. Nat Rev Urol 2020; 17:292-307. [PMID: 32203305 PMCID: PMC7218925 DOI: 10.1038/s41585-020-0298-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
Over the past 5 years, the advent of combination therapeutic strategies has substantially reshaped the clinical management of patients with advanced prostate cancer. However, most of these combination regimens were developed empirically and, despite offering survival benefits, are not enough to halt disease progression. Thus, the development of effective therapeutic strategies that target the mechanisms involved in the acquisition of drug resistance and improve clinical trial design are an unmet clinical need. In this context, we hypothesize that the tumour engineers a dynamic response through the process of cellular rewiring, in which it adapts to the therapy used and develops mechanisms of drug resistance via downstream signalling of key regulatory cascades such as the androgen receptor, PI3K-AKT or GATA2-dependent pathways, as well as initiation of biological processes to revert tumour cells to undifferentiated aggressive states via phenotype switching towards a neuroendocrine phenotype or acquisition of stem-like properties. These dynamic responses are specific for each patient and could be responsible for treatment failure despite multi-target approaches. Understanding the common stages of these cellular rewiring mechanisms to gain a new perspective on the molecular underpinnings of drug resistance might help formulate novel combination therapeutic regimens.
Collapse
Affiliation(s)
- Marc Carceles-Cordon
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - W Kevin Kelly
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leonard Gomella
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karen E Knudsen
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Veronica Rodriguez-Bravo
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Josep Domingo-Domenech
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
15
|
The Role of Crosstalk between AR3 and E2F1 in Drug Resistance in Prostate Cancer Cells. Cells 2020; 9:cells9051094. [PMID: 32354165 PMCID: PMC7290672 DOI: 10.3390/cells9051094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 01/20/2023] Open
Abstract
Background: Drug resistance is one of the most prevalent causes of death in advanced prostate cancer patients. Combination therapies that target cancer cells via different mechanisms to overcome resistance have gained increased attention in recent years. However, the optimal drug combinations and the underlying mechanisms are yet to be fully explored. Aim and methods: The aim of this study is to investigate drug combinations that inhibit the growth of drug-resistant cells and determine the underlying mechanisms of their actions. In addition, we also established cell lines that are resistant to combination treatments and tested new compounds to overcome the phenomenon of double drug-resistance. Results: Our results show that the combination of enzalutamide (ENZ) and docetaxel (DTX) effectively inhibit the growth of prostate cancer cells that are resistant to either drug alone. The downregulation of transcription factor E2F1 plays a crucial role in cellular inhibition in response to the combined therapy. Notably, we found that the androgen receptor (AR) variant AR3 (a.k.a. AR-V7), but not AR full length (AR-FL), positively regulates E2F1 expression in these cells. E2F1 in turn regulates AR3 and forms a positive regulatory feedforward loop. We also established double drug-resistant cell lines that are resistant to ENZ+DTX combination therapy and found that the expression of both AR3 and E2F1 was restored in these cells. Furthermore, we identified that auranofin, an FDA-approved drug for the treatment of rheumatoid arthritis, overcame drug resistance and inhibited the growth of drug-resistant prostate cancer cells both in vitro and in vivo. Conclusion and significance: This proof-of-principle study demonstrates that targeting the E2F1/AR3 feedforward loop via a combination therapy or a multi-targeting drug could circumvent castration resistance in prostate cancer.
Collapse
|
16
|
Yu C, Hu K, Nguyen D, Wang ZA. From genomics to functions: preclinical mouse models for understanding oncogenic pathways in prostate cancer. Am J Cancer Res 2019; 9:2079-2102. [PMID: 31720076 PMCID: PMC6834478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/10/2019] [Indexed: 06/10/2023] Open
Abstract
Next-generation sequencing has revealed numerous genomic alterations that induce aberrant signaling activities in prostate cancer (PCa). Among them are pathways affecting multiple cancer types, including the PI3K/AKT/mTOR, p53, Rb, Ras/Raf/MAPK, Myc, FGF, and Wnt signaling pathways, as well as ones that are prominent in PCa, including alterations in genes of AR signaling, the ETS family, NKX3.1, and SPOP. Cross talk among the oncogenic pathways can confer PCa resistance to therapy, particularly in advanced tumors, which are castration-resistant or show neuroendocrine features. Various experimental models, such as cancer cell lines, animal models, and patient-derived xenografts and organoids have been utilized to dissect PCa progression mechanisms. Here, we review the current preclinical mouse models for studying the most commonly altered pathways in PCa, with an emphasis on their interplays. We highlight the power of genetically engineered mouse models (GEMMs) in translating genomic discoveries into understanding of the functions of these oncogenic events in vivo. Developing and analyzing PCa mouse models will undoubtedly continue to offer new insights into tumor biology and guide novel rationalized therapy.
Collapse
Affiliation(s)
- Chuan Yu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Kevin Hu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Daniel Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| |
Collapse
|
17
|
Ambra1 induces autophagy and desensitizes human prostate cancer cells to cisplatin. Biosci Rep 2019; 39:BSR20170770. [PMID: 29101240 PMCID: PMC6706594 DOI: 10.1042/bsr20170770] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 10/28/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa), the second most mortal cancer from developed countries, presents a high level of chemoresistance. There is emerging evidence underscores the critical role of autophagy in the onset, progression, and chemoresistance of PCa. In the present study, we investigated the possible role of a novel autophagy regulator, activating molecule in beclin1-regulated autophagy1 (Ambra1), a novel ATG gene in the sensitivity or PCa cells to cisplatin. We explored the regulation by the Ambra1 manipulation on the induction of apoptosis and autophagy in human PCa DU145 cells in the presence of cisplatin, via up- or down-regulating Ambra1 expression. In addition, we examined the colony forming of DU145 cells post cisplatin treatment and Ambra1 manipulation. Our results demonstrated that the Ambra1 up-regulation reduced, whereas Ambra1 knockdown increased the cisplatin-induced apoptosis, caspase 3 cleavage, and poly ADP-ribose polymerase (PARP) cleavage. Interestingly, we also found significant autophagy induction in the cisplatin-treated DU145 cells, with increased autophagic vesicles, up-regulated autophagy-related markers. However, the cisplatin-induced autophagy was up-regulated by the Ambra1 overexpression or was down-regulated by the Ambra1 knockdown. In addition, the colony forming was also positively regulated by Ambra1 in DU145 cells post cisplatin treatment. In conclusion, Ambra1 negatively regulates the cisplatin-induced apoptosis and the cisplatin-mediated growth reduction in DU145 cells, in association with the Ambra1-mediated autophagy promotion. It implies that Ambra1-mediated autophagy might be an important mechanism underlining the sensitivity reduction of PCa cells.
Collapse
|
18
|
Xia N, Cui J, Zhu M, Xing R, Lu Y. Androgen receptor variant 12 promotes migration and invasion by regulating MYLK in gastric cancer. J Pathol 2019; 248:304-315. [PMID: 30737779 DOI: 10.1002/path.5257] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/14/2019] [Accepted: 02/06/2019] [Indexed: 12/30/2022]
Abstract
Androgen receptor (AR) and its variants (AR-Vs) promote tumorigenesis and metastasis in many hormone-related cancers, such as breast, prostate and hepatocellular cancers. However, the expression patterns and underlying molecular mechanisms of AR in gastric cancer (GC) are not fully understood. This study aimed to detect the expression of AR-Vs in GC and explored their role in metastasis of GC. Here, the AR expression form was identified in GC cell lines and tissues by RT-PCR and qPCR. Transwell assays and experimental lung metastasis animal models were used to assess the function of AR in cell migration and invasion. Downstream targets of AR were screened by bioinformatics, and identified by luciferase reporter assays and electrophoretic mobility shift assays. AR-v12 was identified as the main expression form in GC cell lines and tissues. Different from full length of AR, AR-v12 was localized to the nucleus independent of androgen. Upregulation of AR-v12 in primary GC tissues was significantly associated with metastasis. Overexpression of AR-v12 promoted migration and invasion independent of androgen. Knockdown of AR-v12 inhibited migration and invasion in vitro, as well as metastasis in vivo. Furthermore, AR-v12, serving as a transcription factor, promoted metastasis through regulating the promoter activity of MYLK. In AR-v12 overexpressing cells, knockdown of MYLK inhibited cell migration and invasion, while in AR-v12 knocked-down cells, overexpression of MYLK promoted cell migration and invasion. Collectively, our study demonstrates that AR-v12 is highly expressed in GC tissues and promotes migration and invasion through directly regulating MYLK. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Nan Xia
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Jiantao Cui
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Min Zhu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Rui Xing
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China.,Department of Medical Oncology, Beijing Hospital, Beijing 100730, PR China
| |
Collapse
|
19
|
Chen W, Yao G, Zhou K. miR-103a-2-5p/miR-30c-1-3p inhibits the progression of prostate cancer resistance to androgen ablation therapy via targeting androgen receptor variant 7. J Cell Biochem 2019; 120:14055-14064. [PMID: 30963631 DOI: 10.1002/jcb.28680] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/14/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
Abstract
Androgens and androgen receptors are vital factors involved in prostate cancer progression, and androgen ablation therapies are commonly used to treat advanced prostate cancer. However, the acquisition of androgen ablation therapy resistance remains a challenge. Recently, androgen receptor splicing variants lacking the ligand-binding domain have been reported to play a critical role in the acquisition of androgen ablation therapy resistance. In the present study, we revealed that the messenger RNA expression and the protein levels of an androgen receptor variant 7 (AR-V7) were higher in prostate cancer tissue samples and in the AR-positive prostate cancer cell line, VCaP. In contrast, microRNA (miR)-30c-1-3p/miR-103a-2-5p expression was significantly downregulated in tumor tissues and cells. miR-30c-1-3p/miR-103a-2-5p overexpression could inhibit AR-V7 expression, suppress VCaP cell growth, and inhibit AR-V7 downstream factor expression by directly targeting the 3'-untranslated region of AR-V7. Under enzalutamide (Enza) treatment, the effects of AR-V7 overexpression were the opposite of those of miR-103a-2-5p/miR-30c-1-3p overexpression; more importantly, the effects of miR-103a-2-5p/miR-30c-1-3p overexpression could be significantly reversed by AR-V7 overexpression under Enza. In summary, we demonstrated a novel mechanism of the miR-30c-1-3p/miR-103a-2-5p/AR-V7 axis modulating the cell proliferation of AR-positive prostate cancer cells via AR downstream targets. The clinical application of miR-30c-1-3p/miR-103a-2-5p needs further in vivo validation.
Collapse
Affiliation(s)
- Wenjie Chen
- Department of Urology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gan Yao
- Department of Urology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Keqin Zhou
- Department of Urology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Cheaito KA, Bahmad HF, Hadadeh O, Saleh E, Dagher C, Hammoud MS, Shahait M, Mrad ZA, Nassif S, Tawil A, Bulbul M, Khauli R, Wazzan W, Nasr R, Shamseddine A, Temraz S, El-Sabban ME, El-Hajj A, Mukherji D, Abou-Kheir W. EMT Markers in Locally-Advanced Prostate Cancer: Predicting Recurrence? Front Oncol 2019; 9:131. [PMID: 30915272 PMCID: PMC6421270 DOI: 10.3389/fonc.2019.00131] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Prostate cancer (PCa) is the second most frequent cause of cancer-related death in men worldwide. It is a heterogeneous disease at molecular and clinical levels which makes its prognosis and treatment outcome hard to predict. The epithelial-to-mesenchymal transition (EMT) marks a key step in the invasion and malignant progression of PCa. We sought to assess the co-expression of epithelial cytokeratin 8 (CK8) and mesenchymal vimentin (Vim) in locally-advanced PCa as indicators of EMT and consequently predictors of the progression status of the disease. Methods: Co-expression of CK8 and Vim was evaluated by immunofluorescence (IF) on paraffin-embedded tissue sections of 122 patients with PCa who underwent radical prostatectomies between 1998 and 2016 at the American University of Beirut Medical Center (AUBMC). EMT score was calculated accordingly and then correlated with the patients' clinicopathological parameters and PSA failure. Results: The co-expression of CK8/Vim (EMT score), was associated with increasing Gleason group. A highly significant linear association was detected wherein higher Gleason group was associated with higher mean EMT score. In addition, the median estimated biochemical recurrence-free survival for patients with < 25% EMT score was almost double that of patients with more than 25%. The validity of this score for prediction of prognosis was further demonstrated using cox regression model. Our data also confirmed that the EMT score can predict PSA failure irrespective of Gleason group, pathological stage, or surgical margins. Conclusion: This study suggests that assessment of molecular markers of EMT, particularly CK8 and Vim, in radical prostatectomy specimens, in addition to conventional clinicopathological prognostic parameters, can aid in the development of a novel system for predicting the prognosis of locally-advanced PCa.
Collapse
Affiliation(s)
- Katia A Cheaito
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hisham F Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ola Hadadeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Eman Saleh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Christelle Dagher
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Miza Salim Hammoud
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohammad Shahait
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Zaki Abou Mrad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samer Nassif
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ayman Tawil
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Muhammad Bulbul
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Raja Khauli
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Wassim Wazzan
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rami Nasr
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Marwan E El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Albert El-Hajj
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
21
|
Arriaga JM, Abate-Shen C. Genetically Engineered Mouse Models of Prostate Cancer in the Postgenomic Era. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a030528. [PMID: 29661807 DOI: 10.1101/cshperspect.a030528] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Recent genomic sequencing analyses have unveiled the spectrum of genomic alterations that occur in primary and advanced prostate cancer, raising the question of whether the corresponding genes are functionally relevant for prostate tumorigenesis, and whether such functions are associated with particular disease stages. In this review, we describe genetically engineered mouse models (GEMMs) of prostate cancer, focusing on those that model genomic alterations known to occur in human prostate cancer. We consider whether the phenotypes of GEMMs based on gain or loss of function of the relevant genes provide reliable counterparts to study the predicted consequences of the corresponding genomic alterations as occur in human prostate cancer, and we discuss exceptions in which the GEMMs do not fully emulate the expected phenotypes. Last, we highlight future directions for the generation of new GEMMs of prostate cancer and consider how we can use GEMMs most effectively to decipher the biological and molecular mechanisms of disease progression, as well as to tackle clinically relevant questions.
Collapse
Affiliation(s)
- Juan M Arriaga
- Departments of Urology, Medicine, Systems Biology, and Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| | - Cory Abate-Shen
- Departments of Urology, Medicine, Systems Biology, and Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
22
|
Montes M, Sanford BL, Comiskey DF, Chandler DS. RNA Splicing and Disease: Animal Models to Therapies. Trends Genet 2019; 35:68-87. [PMID: 30466729 PMCID: PMC6339821 DOI: 10.1016/j.tig.2018.10.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/01/2018] [Accepted: 10/16/2018] [Indexed: 02/07/2023]
Abstract
Alternative splicing of pre-mRNA increases genetic diversity, and recent studies estimate that most human multiexon genes are alternatively spliced. If this process is not highly regulated and accurate, it leads to mis-splicing events, which may result in proteins with altered function. A growing body of work has implicated mis-splicing events in a range of diseases, including cancer, neurodegenerative diseases, and muscular dystrophies. Understanding the mechanisms that cause aberrant splicing events and how this leads to disease is vital for designing effective therapeutic strategies. In this review, we focus on advances in therapies targeting splicing, and highlight the animal models developed to recapitulate disease phenotypes as a model for testing these therapies.
Collapse
Affiliation(s)
- Matías Montes
- Molecular, Cellular, and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, OH, USA; Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Brianne L Sanford
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Daniel F Comiskey
- Molecular, Cellular, and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, OH, USA; Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Dawn S Chandler
- Molecular, Cellular, and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, OH, USA; Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
23
|
Sharp A, Coleman I, Yuan W, Sprenger C, Dolling D, Rodrigues DN, Russo JW, Figueiredo I, Bertan C, Seed G, Riisnaes R, Uo T, Neeb A, Welti J, Morrissey C, Carreira S, Luo J, Nelson PS, Balk SP, True LD, de Bono JS, Plymate SR. Androgen receptor splice variant-7 expression emerges with castration resistance in prostate cancer. J Clin Invest 2018; 129:192-208. [PMID: 30334814 PMCID: PMC6307949 DOI: 10.1172/jci122819] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/09/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Liquid biopsies have demonstrated that the constitutively active androgen receptor splice variant-7 (AR-V7) associates with reduced response and overall survival from endocrine therapies in castration-resistant prostate cancer (CRPC). However, these studies provide little information pertaining to AR-V7 expression in prostate cancer (PC) tissue. METHODS Following generation and validation of a potentially novel AR-V7 antibody for IHC, AR-V7 protein expression was determined for 358 primary prostate samples and 293 metastatic biopsies. Associations with disease progression, full-length androgen receptor (AR-FL) expression, response to therapy, and gene expression were determined. RESULTS We demonstrated that AR-V7 protein is rarely expressed (<1%) in primary PC but is frequently detected (75% of cases) following androgen deprivation therapy, with further significant (P = 0.020) increase in expression following abiraterone acetate or enzalutamide therapy. In CRPC, AR-V7 expression is predominantly (94% of cases) nuclear and correlates with AR-FL expression (P ≤ 0.001) and AR copy number (P = 0.026). However, dissociation of expression was observed, suggesting that mRNA splicing remains crucial for AR-V7 generation. AR-V7 expression was heterogeneous between different metastases from a patient, although AR-V7 expression was similar within a metastasis. Moreover, AR-V7 expression correlated with a unique 59-gene signature in CRPC, including HOXB13, a critical coregulator of AR-V7 function. Finally, AR-V7-negative disease associated with better prostate-specific antigen (PSA) responses (100% vs. 54%, P = 0.03) and overall survival (74.3 vs. 25.2 months, hazard ratio 0.23 [0.07-0.79], P = 0.02) from endocrine therapies (pre-chemotherapy). CONCLUSION This study provides impetus to develop therapies that abrogate AR-V7 signaling to improve our understanding of AR-V7 biology and to confirm the clinical significance of AR-V7. FUNDING Work at the University of Washington and in the Plymate and Nelson laboratories is supported by the Department of Defense Prostate Cancer Research Program (W81XWH-14-2-0183, W81XWH-12-PCRP-TIA, W81XWH-15-1-0430, and W81XWH-13-2-0070), the Pacific Northwest Prostate Cancer SPORE (P50CA97186), the Institute for Prostate Cancer Research, the Veterans Affairs Research Program, the NIH/National Cancer Institute (P01CA163227), and the Prostate Cancer Foundation. Work in the de Bono laboratory was supported by funding from the Movember Foundation/Prostate Cancer UK (CEO13-2-002), the US Department of Defense (W81XWH-13-2-0093), the Prostate Cancer Foundation (20131017 and 20131017-1), Stand Up To Cancer (SU2C-AACR-DT0712), Cancer Research UK (CRM108X-A25144), and the UK Department of Health through an Experimental Cancer Medicine Centre grant (ECMC-CRM064X).
Collapse
Affiliation(s)
- Adam Sharp
- The Institute of Cancer Research, London, United Kingdom.,The Royal Marsden, London, United Kingdom
| | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Wei Yuan
- The Institute of Cancer Research, London, United Kingdom
| | - Cynthia Sprenger
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - David Dolling
- The Institute of Cancer Research, London, United Kingdom
| | | | - Joshua W Russo
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Claudia Bertan
- The Institute of Cancer Research, London, United Kingdom
| | - George Seed
- The Institute of Cancer Research, London, United Kingdom
| | - Ruth Riisnaes
- The Institute of Cancer Research, London, United Kingdom
| | - Takuma Uo
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Antje Neeb
- The Institute of Cancer Research, London, United Kingdom
| | - Jonathan Welti
- The Institute of Cancer Research, London, United Kingdom
| | - Colm Morrissey
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Jun Luo
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Steven P Balk
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Lawrence D True
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Johann S de Bono
- The Institute of Cancer Research, London, United Kingdom.,The Royal Marsden, London, United Kingdom
| | - Stephen R Plymate
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Puget Sound VA Health Care System, Geriatric Research Education and Clinical Center (PSVAHCS-GRECC), Seattle, Washington, USA
| |
Collapse
|
24
|
Scarano WR, Pinho CF, Pissinatti L, Gonçalves BF, Mendes LO, Campos SG. Cell junctions in the prostate: an overview about the effects of Endocrine Disrupting Chemicals (EDCS) in different experimental models. Reprod Toxicol 2018; 81:147-154. [DOI: 10.1016/j.reprotox.2018.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/30/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
|
25
|
Androgen Receptor Variants and Castration-resistant Prostate Cancer: Looking Back and Looking Forward. Eur Urol 2018; 73:724-726. [DOI: 10.1016/j.eururo.2017.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022]
|
26
|
Davies AH, Beltran H, Zoubeidi A. Cellular plasticity and the neuroendocrine phenotype in prostate cancer. Nat Rev Urol 2018; 15:271-286. [PMID: 29460922 DOI: 10.1038/nrurol.2018.22] [Citation(s) in RCA: 291] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The success of next-generation androgen receptor (AR) pathway inhibitors, such as abiraterone acetate and enzalutamide, in treating prostate cancer has been hampered by the emergence of drug resistance. This acquired drug resistance is driven, in part, by the ability of prostate cancer cells to change their phenotype to adopt AR-independent pathways for growth and survival. Around one-quarter of resistant prostate tumours comprise cells that have undergone cellular reprogramming to become AR-independent and to acquire a continuum of neuroendocrine characteristics. These highly aggressive and lethal tumours, termed neuroendocrine prostate cancer (NEPC), exhibit reactivation of developmental programmes that are associated with epithelial-mesenchymal plasticity and acquisition of stem-like cell properties. In the past few years, our understanding of the link between lineage plasticity and an emergent NEPC phenotype has considerably increased. This new knowledge can contribute to novel therapeutic modalities that are likely to improve the treatment and clinical management of aggressive prostate cancer.
Collapse
Affiliation(s)
- Alastair H Davies
- Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, BC, Canada
| | - Himisha Beltran
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, 413 East 69th Street, New York, NY, USA
| | - Amina Zoubeidi
- Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, BC, Canada
| |
Collapse
|
27
|
Lo UG, Lee CF, Lee MS, Hsieh JT. The Role and Mechanism of Epithelial-to-Mesenchymal Transition in Prostate Cancer Progression. Int J Mol Sci 2017; 18:ijms18102079. [PMID: 28973968 PMCID: PMC5666761 DOI: 10.3390/ijms18102079] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/21/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
In prostate cancer (PCa), similar to many other cancers, distant organ metastasis symbolizes the beginning of the end disease, which eventually leads to cancer death. Many mechanisms have been identified in this process that can be rationalized into targeted therapy. Among them, epithelial-to-mesenchymal transition (EMT) is originally characterized as a critical step for cell trans-differentiation during embryo development and now recognized in promoting cancer cells invasiveness because of high mobility and migratory abilities of mesenchymal cells once converted from carcinoma cells. Nevertheless, the underlying pathways leading to EMT appear to be very diverse in different cancer types, which certainly represent a challenge for developing effective intervention. In this article, we have carefully reviewed the key factors involved in EMT of PCa with clinical correlation in hope to facilitate the development of new therapeutic strategy that is expected to reduce the disease mortality.
Collapse
Affiliation(s)
- U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Cheng-Fan Lee
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
28
|
Schreyer E, Barthélémy P, Cottard F, Ould Madi-Berthélémy P, Schaff-Wendling F, Kurtz JE, Céraline J. [Androgen receptor variants in prostate cancer]. Med Sci (Paris) 2017; 33:758-764. [PMID: 28945566 DOI: 10.1051/medsci/20173308021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Prostate cancer is a public health concern as it currently represents the most frequent malignancy in men in Europe. Progression of this hormone-dependent cancer is driven by androgens. Thus, the most common treatment for patients with advanced prostate cancer consists in an androgen ablation by castration therapy. However, the majority of patients relapses and develops a castration-resistant prostate cancer. This failure of androgen deprivation is related to the emergence of mutant and splice variants of the androgen receptor. Indeed, androgen receptor variants are ligand-independent, constitutively active and thus able to induce resistance to castration. This review focuses on AR variants signaling pathways and their role in resistance to castration and prostate cancer progression.
Collapse
Affiliation(s)
- Edwige Schreyer
- Université de Strasbourg, Inserm, VSDSC UMR-S 1113, IGBMC, 1, rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France
| | - Philippe Barthélémy
- Université de Strasbourg, Inserm, VSDSC UMR-S 1113, IGBMC, 1, rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France - Service d'oncologie et d'hématologie, Hôpitaux universitaires de Strasbourg, 67000 Strasbourg, France
| | - Félicie Cottard
- Department of urology, Center for Clinical research, University Freiburg Medical Center, Breisacherstrasse 66, D-79106 Freiburg, Allemagne
| | - Pauline Ould Madi-Berthélémy
- Université de Strasbourg, Inserm, VSDSC UMR-S 1113, IGBMC, 1, rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France
| | - Frédérique Schaff-Wendling
- Université de Strasbourg, Inserm, VSDSC UMR-S 1113, IGBMC, 1, rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France - Service d'oncologie et d'hématologie, Hôpitaux universitaires de Strasbourg, 67000 Strasbourg, France
| | - Jean-Emmanuel Kurtz
- Université de Strasbourg, Inserm, VSDSC UMR-S 1113, IGBMC, 1, rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France - Service d'oncologie et d'hématologie, Hôpitaux universitaires de Strasbourg, 67000 Strasbourg, France
| | - Jocelyn Céraline
- Université de Strasbourg, Inserm, VSDSC UMR-S 1113, IGBMC, 1, rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France - Service d'oncologie et d'hématologie, Hôpitaux universitaires de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
29
|
Munkley J, Livermore K, Rajan P, Elliott DJ. RNA splicing and splicing regulator changes in prostate cancer pathology. Hum Genet 2017; 136:1143-1154. [PMID: 28382513 PMCID: PMC5602090 DOI: 10.1007/s00439-017-1792-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/29/2017] [Indexed: 11/26/2022]
Abstract
Changes in mRNA splice patterns have been associated with key pathological mechanisms in prostate cancer progression. The androgen receptor (abbreviated AR) transcription factor is a major driver of prostate cancer pathology and activated by androgen steroid hormones. Selection of alternative promoters by the activated AR can critically alter gene function by switching mRNA isoform production, including creating a pro-oncogenic isoform of the normally tumour suppressor gene TSC2. A number of androgen-regulated genes generate alternatively spliced mRNA isoforms, including a prostate-specific splice isoform of ST6GALNAC1 mRNA. ST6GALNAC1 encodes a sialyltransferase that catalyses the synthesis of the cancer-associated sTn antigen important for cell mobility. Genetic rearrangements occurring early in prostate cancer development place ERG oncogene expression under the control of the androgen-regulated TMPRSS2 promoter to hijack cell behaviour. This TMPRSS2-ERG fusion gene shows different patterns of alternative splicing in invasive versus localised prostate cancer. Alternative AR mRNA isoforms play a key role in the generation of prostate cancer drug resistance, by providing a mechanism through which prostate cancer cells can grow in limited serum androgen concentrations. A number of splicing regulator proteins change expression patterns in prostate cancer and may help drive key stages of disease progression. Up-regulation of SRRM4 establishes neuronal splicing patterns in neuroendocrine prostate cancer. The splicing regulators Sam68 and Tra2β increase expression in prostate cancer. The SR protein kinase SRPK1 that modulates the activity of SR proteins is up-regulated in prostate cancer and has already given encouraging results as a potential therapeutic target in mouse models.
Collapse
Affiliation(s)
- Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle, NE1 3BZ, England, UK
| | - Karen Livermore
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle, NE1 3BZ, England, UK
| | - Prabhakar Rajan
- Barts Cancer Institute, John Vane Science Centre, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - David J Elliott
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle, NE1 3BZ, England, UK.
| |
Collapse
|
30
|
Chen WY, Tsai YC, Yeh HL, Suau F, Jiang KC, Shao AN, Huang J, Liu YN. Loss of SPDEF and gain of TGFBI activity after androgen deprivation therapy promote EMT and bone metastasis of prostate cancer. Sci Signal 2017; 10:10/492/eaam6826. [PMID: 28811384 DOI: 10.1126/scisignal.aam6826] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Androgen deprivation therapy (ADT) targeting the androgen receptor (AR) is a standard therapeutic regimen for treating prostate cancer. However, most tumors progress to metastatic castration-resistant prostate cancer after ADT. We identified the type 1, 2, and 4 collagen-binding protein transforming growth factor-β (TGFβ)-induced protein (TGFBI) as an important factor in the epithelial-to-mesenchymal transition (EMT) and malignant progression of prostate cancer. In prostate cancer cell lines, AR signaling stimulated the activity of the transcription factor SPDEF, which repressed the expression of TGFBI ADT, AR antagonism, or overexpression of TGFBI inhibited the activity of SPDEF and enhanced the proliferation rates of prostate cancer cells. Knockdown of TGFBI suppressed migration and proliferation in cultured cells and reduced prostate tumor growth and brain and bone metastasis in xenograft models, extending the survival of tumor-bearing mice. Analysis of prostate tissue samples collected before and after ADT from the same patients showed that ADT reduced the nuclear abundance of SPDEF and increased the production of TGFBI. Our findings suggest that induction of TGFBI promotes prostate cancer growth and metastasis and can be caused by dysregulation or therapeutic inhibition of AR signaling.
Collapse
Affiliation(s)
- Wei-Yu Chen
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan.,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yuan-Chin Tsai
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Lien Yeh
- Institute of Information System and Applications, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Florent Suau
- Department of Microbiology, Faculty of Pharmacy, Dicle University, Diyarbakir 21280, Turkey
| | - Kuo-Ching Jiang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ai-Ning Shao
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Jiaoti Huang
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
31
|
Cottard F, Madi-Berthélémy PO, Erdmann E, Schaff-Wendling F, Keime C, Ye T, Kurtz JE, Céraline J. Dual effects of constitutively active androgen receptor and full-length androgen receptor for N-cadherin regulation in prostate cancer. Oncotarget 2017; 8:72008-72020. [PMID: 29069764 PMCID: PMC5641107 DOI: 10.18632/oncotarget.18270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 05/12/2017] [Indexed: 12/18/2022] Open
Abstract
Constitutively active androgen receptor (AR) variants have been involved in the expression of mesenchymal markers such as N-cadherin in prostate cancer (PCa). However, the underlying molecular mechanisms remain elusive. It remains unclear, whether N-cadherin gene (CDH2) is a direct transcriptional target of AR variants or whether the observed upregulation is due to indirect effects through additional regulatory factors. Moreover, the specific contribution of full-length AR and AR variants in N-cadherin regulation in PCa has never been explored deeply. To investigate this, we artificially mimicked the co-expression of AR variants together with a full-length AR and performed miRNA-seq, RNA-seq and ChIP assays. Our results were in favor of a direct AR variants action on CDH2. Our data also revealed a distinctive mode of action between full-length AR and AR variants to regulate N-cadherin expression. Both wild type AR and AR variants could interact with a regulatory element in intron 1 of CDH2. However, a higher histone H4 acetylation in this genomic region was only observed with AR variants. This suggests that full-length AR may play an occluding function to impede CDH2 upregulation. Our data further highlighted a negative effect of AR variants on the expression of the endogenous full-length AR in LNCaP. These differences in the mode of action of AR variants and full-length AR for the control of one key gene for prostate cancer progression could be worth considering for targeting AR variants in PCa.
Collapse
Affiliation(s)
| | | | - Eva Erdmann
- Université de Strasbourg, INSERM, FMTS, Strasbourg, France
| | - Frédérique Schaff-Wendling
- Université de Strasbourg, INSERM, FMTS, Strasbourg, France.,Service d'Onco-Hématologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Céline Keime
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, France
| | - Tao Ye
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, France
| | - Jean-Emmanuel Kurtz
- Université de Strasbourg, INSERM, FMTS, Strasbourg, France.,Service d'Onco-Hématologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jocelyn Céraline
- Université de Strasbourg, INSERM, FMTS, Strasbourg, France.,Service d'Onco-Hématologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
32
|
Wadosky KM, Koochekpour S. Androgen receptor splice variants and prostate cancer: From bench to bedside. Oncotarget 2017; 8:18550-18576. [PMID: 28077788 PMCID: PMC5392349 DOI: 10.18632/oncotarget.14537] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/31/2016] [Indexed: 12/13/2022] Open
Abstract
Therapeutic interventions for advanced prostate cancer (PCa) center on inhibiting androgen receptor (AR) and downstream signaling pathways. Resistance to androgen deprivation therapy and/or AR antagonists is inevitable and molecular mechanisms driving castration-resistant PCa (CR-PCa) primarily involve alterations in AR expression and activity. Detailed molecular biology work over the past decade, discussed at length in this review article, has revealed several AR transcripts that result from alternative splicing. These AR splice variants are increased in cell and mouse models of CR-PCa and in CR-PCa tumors. Several AR variants lack the ligand binding domain, but retain their ability to bind DNA and activate transcription-linking constitutive AR function and therapeutic failure. ARV7 is the only variant endogenously detected at the protein level and thus has undergone more thorough molecular characterization. Clinical trials in PCa are currently investigating ARV7 utility as a biomarker and new therapeutics that inhibit ARV7 . Overall, this review will illustrate the historical perspectives of AR splice variant discovery using fundamental molecular biology techniques and how it changed the clinical approach to both therapeutic decisions and strategy. The body of work investigating AR splice variants in PCa represents a true example of translational research from bench to bedside.
Collapse
Affiliation(s)
- Kristine M. Wadosky
- Department of Cancer Genetics, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Shahriar Koochekpour
- Department of Cancer Genetics, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Urology, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
33
|
Li Y, Zhang DJ, Qiu Y, Kido T, Lau YFC. The Y-located proto-oncogene TSPY exacerbates and its X-homologue TSPX inhibits transactivation functions of androgen receptor and its constitutively active variants. Hum Mol Genet 2017; 26:901-912. [PMID: 28169398 PMCID: PMC6075507 DOI: 10.1093/hmg/ddx005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/05/2016] [Accepted: 01/03/2017] [Indexed: 12/15/2022] Open
Abstract
The gonadoblastoma gene, testis-specific protein Y-encoded (TSPY), on the Y chromosome and its X-homologue, TSPX, are cell cycle regulators and function as a proto-oncogene and a tumor suppressor respectively in human oncogenesis. TSPY and TSPX competitively bind to the androgen receptor (AR) and AR variants, such as AR-V7, at their conserved SET/NAP domain, and exacerbate and repress the transactivation of the AR/AR-V7 target genes in ligand dependent and independent manners respectively. The inhibitory domain has been mapped to the carboxyl acidic domain of TSPX, truncation of which renders TSPX to be stimulatory while its transposition to the C-terminus of TSPY results in an inhibitory hybrid protein. TSPY and TSPX co-localize with the endogenous AR, in the presence of ligand, on the promoters and differentially regulate the expression of the endogenous AR target genes in the androgen-responsive LNCaP prostate cancer cells. Transcriptome analysis shows that TSPY and TSPX expressions differentially affect significant numbers of canonical pathways, upstream regulators and cellular functions. Significantly, among the common ones, TSPY activates and TSPX inhibits numerous growth-related and oncogenic canonical pathways and cellular functions in the respective cell populations. Hence, TSPY and TSPX exert opposing effects on the transactivation functions of AR and AR-Vs important for various physiological and disease processes sensitive to male sex hormone actions, thereby not only affecting the pathogenesis of male-specific prostate cancer but also likely contributing to sex differences in the health and diseases of man.
Collapse
Affiliation(s)
- Yunmin Li
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center
- Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | - Dong Ji Zhang
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center
- Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | - Yun Qiu
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Tatsuo Kido
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center
- Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| | - Yun-Fai Chris Lau
- Division of Cell and Developmental Genetics, Department of Medicine, VA Medical Center
- Institute for Human Genetics, University of California, San Francisco, CA 94121, USA
| |
Collapse
|
34
|
Mizokami A, Izumi K, Konaka H, Kitagawa Y, Kadono Y, Narimoto K, Nohara T, Bahl AK, Namiki M. Understanding prostate-specific antigen dynamics in monitoring metastatic castration-resistant prostate cancer: implications for clinical practice. Asian J Androl 2017; 19:143-148. [PMID: 27270339 PMCID: PMC5312209 DOI: 10.4103/1008-682x.179159] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Availability of novel hormonal therapies as well as docetaxel and cabazitaxel treatment for metastatic castration-resistant prostate cancer (CRPC) has changed the outlook for this group of patients with improvements in progression-free survival and overall survival. Physicians often diagnose the progression of prostate cancer using serum prostate-specific antigen (PSA). However, serum PSA is not always correlated with the clinical status in CRPC. To evaluate the PSA dynamics with greater precision, understanding of the control of PSA and of the mechanisms of development of CRPC is needed. Moreover, it is necessary to use new hormonal therapies with an appropriate timing to optimally improve the prognosis and the QOL of the patients. In the present review, we ascertain the PSA dynamics and the mechanisms of the development of CRPC to assist in optimal utilization of the new treatments for mCRPC.
Collapse
Affiliation(s)
- Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| | - Kouji Izumi
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| | - Hiroyuki Konaka
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| | - Yasuhide Kitagawa
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| | - Yoshifumi Kadono
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| | - Kazutaka Narimoto
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| | - Takahiro Nohara
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| | - Amit K Bahl
- Bristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, BS2 8ED, UK
| | - Mikio Namiki
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, 13-1 Takaramachi Kanazawa, 920-8640 Japan
| |
Collapse
|
35
|
Scher HI, Lu D, Schreiber NA, Louw J, Graf RP, Vargas HA, Johnson A, Jendrisak A, Bambury R, Danila D, McLaughlin B, Wahl J, Greene SB, Heller G, Marrinucci D, Fleisher M, Dittamore R. Association of AR-V7 on Circulating Tumor Cells as a Treatment-Specific Biomarker With Outcomes and Survival in Castration-Resistant Prostate Cancer. JAMA Oncol 2017; 2:1441-1449. [PMID: 27262168 DOI: 10.1001/jamaoncol.2016.1828] [Citation(s) in RCA: 519] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Importance A critical decision in the management of metastatic castration-resistant prostate cancer (mCRPC) is when to administer an androgen receptor signaling (ARS) inhibitor or a taxane. Objective To determine if pretherapy nuclear androgen-receptor splice variant 7 (AR-V7) protein expression and localization on circulating tumor cells (CTCs) is a treatment-specific marker for response and outcomes between ARS inhibitors and taxanes. Design, Setting, and Participants For this cross-sectional cohort study at Memorial Sloan Kettering Cancer Center, 265 men with progressive mCRPC undergoing a change in treatment were considered; 86 were excluded because they were not initiating ARS or taxane therapy; and 18 were excluded for processing time constraints, leaving 161 patients for analysis. Between December 2012 and March 2015, blood was collected and processed from patients with progressive mCRPC immediately prior to new line of systemic therapy. Patients were followed up to 3 years. Main Outcomes and Measures Prostate-specific antigen (PSA) response, time receiving therapy, radiographic progression-free survival (rPFS), and overall survival (OS). Results Overall, of 193 prospectively collected blood samples from 161 men with mCRPC, 191 were evaluable (128 pre-ARS inhibitor and 63 pretaxane). AR-V7-positive CTCs were found in 34 samples (18%), including 3% of first-line, 18% of second-line, and 31% of third- or greater line samples. Patients whose samples had AR-V7-positive CTCs before ARS inhibition had resistant posttherapy PSA changes (PTPC), shorter rPFS, shorter time on therapy, and shorter OS than those without AR-V7-positive CTCs. Overall, resistant PTPC were seen in 65 of 112 samples (58%) without detectable AR-V7-positive CTCs prior to ARS inhibition. There were statistically significant differences in OS but not in PTPC, time on therapy, or rPFS for patients with or without pretherapy AR-V7-positive CTCs treated with a taxane. A multivariable model adjusting for baseline factors associated with survival showed superior OS with taxanes relative to ARS inhibitors when AR-V7-positive CTCs were detected pretherapy (hazard ratio, 0.24; 95% CI, 0.10-0.57; P = .035). Conclusions and Relevance The results validate CTC nuclear expression of AR-V7 protein in men with mCRPC as a treatment-specific biomarker that is associated with superior survival on taxane therapy over ARS-directed therapy in a clinical practice setting. Continued examination of this biomarker in prospective studies will further aid clinical utility.
Collapse
Affiliation(s)
- Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York2Department of Medicine, Weill Cornell Medical College, New York, New York
| | - David Lu
- Epic Sciences, La Jolla, California
| | - Nicole A Schreiber
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - Hebert A Vargas
- Body Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York5Department of Radiology, Weill Cornell Medical College, New York, New York
| | | | | | - Richard Bambury
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York6Cancer Services, Department of Medical Oncology, Cork University Hospital, Wilton, Cork, Ireland
| | - Daniel Danila
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York2Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Brigit McLaughlin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - Glenn Heller
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Martin Fleisher
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
36
|
Azoitei A, Merseburger AS, Godau B, Hoda MR, Schmid E, Cronauer MV. C-terminally truncated constitutively active androgen receptor variants and their biologic and clinical significance in castration-resistant prostate cancer. J Steroid Biochem Mol Biol 2017; 166:38-44. [PMID: 27345700 DOI: 10.1016/j.jsbmb.2016.06.008] [Citation(s) in RCA: 14] [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: 02/05/2016] [Revised: 06/10/2016] [Accepted: 06/20/2016] [Indexed: 10/21/2022]
Abstract
A mechanism allowing castration resistant prostate cancer cells to escape the effects of conventional anti-hormonal treatments is the synthesis of constitutively active, C-terminally truncated androgen receptor (AR)-variants. Lacking the entire or vast parts of the ligand binding domain, the intended target of traditional endocrine therapies, these AR-variants (termed ARΔLBD) are insensitive to all traditional treatments including second generation compounds like abiraterone, enzalutamide or ARN-509. Although ARΔLBD are predominantly products of alternative splicing, they can also be products of nonsense mutations or proteolytic cleavage. In this review, we will discuss the etiology and function of c-terminally truncated AR-variants and their clinical significance as markers/targets for the treatment of castration resistant prostate cancer.
Collapse
Affiliation(s)
- Anca Azoitei
- Department of Urology, Ulm University Medical School, 89075 Ulm, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Beate Godau
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - M Raschid Hoda
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Evi Schmid
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Marcus V Cronauer
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany.
| |
Collapse
|
37
|
Scher HI, Graf RP, Schreiber NA, McLaughlin B, Lu D, Louw J, Danila DC, Dugan L, Johnson A, Heller G, Fleisher M, Dittamore R. Nuclear-specific AR-V7 Protein Localization is Necessary to Guide Treatment Selection in Metastatic Castration-resistant Prostate Cancer. Eur Urol 2016; 71:874-882. [PMID: 27979426 DOI: 10.1016/j.eururo.2016.11.024] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/16/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Circulating tumor cells (CTCs) expressing AR-V7 protein localized to the nucleus (nuclear-specific) identify metastatic castration-resistant prostate cancer (mCRPC) patients with improved overall survival (OS) on taxane therapy relative to the androgen receptor signaling inhibitors (ARSi) abiraterone acetate, enzalutamide, and apalutamide. OBJECTIVE To evaluate if expanding the positivity criteria to include both nuclear and cytoplasmic AR-V7 localization ("nuclear-agnostic") identifies more patients who would benefit from a taxane over an ARSi. DESIGN, SETTING, AND PARTICIPANTS The study used a cross-sectional cohort. Between December 2012 and March 2015, 193 pretherapy blood samples, 191 of which were evaluable, were collected and processed from 161 unique mCRPC patients before starting a new line of systemic therapy for disease progression at the Memorial Sloan Kettering Cancer Center. The association between two AR-V7 scoring criteria, post-therapy prostate-specific antigen (PSA) change (PTPC) and OS following ARSi or taxane treatment, was explored. One criterion required nuclear-specific AR-V7 localization, and the other required an AR-V7 signal but was agnostic to protein localization in CTCs. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSES Correlation of AR-V7 status to PTPC and OS was investigated. Relationships with survival were analyzed using multivariable Cox regression and log-rank analyses. RESULTS AND LIMITATIONS A total of 34 (18%) samples were AR-V7-positive using nuclear-specific criteria, and 56 (29%) were AR-V7-positive using nuclear-agnostic criteria. Following ARSi treatment, none of the 16 nuclear-specific AR-V7-positive samples and six of the 32 (19%) nuclear-agnostic AR-V7-positive samples had ≥50% PTPC at 12 weeks. The strongest baseline factor influencing OS was the interaction between the presence of nuclear-specific AR-V7-positive CTCs and treatment with a taxane (hazard ratio 0.24, 95% confidence interval 0.078-0.79; p=0.019). This interaction was not significant when nuclear-agnostic criteria were used. CONCLUSIONS To reliably inform treatment selection using an AR-V7 protein biomarker in CTCs, nuclear-specific localization is required. PATIENT SUMMARY We analyzed outcomes for patients with metastatic castration-resistant prostate cancer on androgen receptor signaling inhibitors and standard chemotherapy. Patients with circulating tumor cells that had AR-V7 protein in the cellular nuclei were very likely to survive longer on taxane-based chemotherapy, and tests unable to distinguish where the protein is located in the cell are not as predictive of benefit.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Androgen Antagonists/therapeutic use
- Antineoplastic Agents, Phytogenic/therapeutic use
- Biomarkers, Tumor/blood
- Cell Nucleus/chemistry
- Cell Nucleus/pathology
- Chi-Square Distribution
- Cross-Sectional Studies
- Humans
- Kaplan-Meier Estimate
- Liquid Biopsy
- Male
- Middle Aged
- Multivariate Analysis
- Neoplastic Cells, Circulating/chemistry
- Neoplastic Cells, Circulating/pathology
- Patient Selection
- Predictive Value of Tests
- Prognosis
- Proportional Hazards Models
- Prostatic Neoplasms, Castration-Resistant/blood
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/mortality
- Prostatic Neoplasms, Castration-Resistant/pathology
- Protein Isoforms
- Receptors, Androgen/blood
- Taxoids/therapeutic use
- Time Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | | | - Nicole A Schreiber
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brigit McLaughlin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Lu
- Epic Sciences, La Jolla, CA, USA
| | | | - Daniel C Danila
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | | | - Glenn Heller
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin Fleisher
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
38
|
Liu X, Ledet E, Li D, Dotiwala A, Steinberger A, Feibus A, Li J, Qi Y, Silberstein J, Lee B, Dong Y, Sartor O, Zhang H. A Whole Blood Assay for AR-V7 and AR v567es in Patients with Prostate Cancer. J Urol 2016; 196:1758-1763. [PMID: 27449259 PMCID: PMC5161406 DOI: 10.1016/j.juro.2016.06.095] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2016] [Indexed: 01/18/2023]
Abstract
PURPOSE Most prostate cancer mortality can be attributed to metastatic castration resistant prostate cancer, an advanced stage that remains incurable despite recent advances. The AR (androgen receptor) signaling axis remains active in castration resistant prostate cancer. Recent studies suggest that expression of the AR-V (AR splice variant) AR-V7 may underlie resistance to abiraterone and enzalutamide. However, controversy exists over the optimal assay. Our objective was to develop a fast and sensitive assay for AR-Vs in patients. MATERIALS AND METHODS Two approaches were assessed in this study. The first approach was based on depletion of leukocytes and the second one used RNA purified directly from whole blood preserved in PAXgene® tubes. Transcript expression was analyzed by quantitative reverse transcription-polymerase chain reaction. RESULTS Through a side-by-side comparison we found that the whole blood approach was suitable to detect AR-Vs. The specificity of the assay was corroborated in a cancer-free cohort. Using the PAXgene assay samples from a cohort of 46 patients with castration resistant prostate cancer were analyzed. Overall, AR-V7 and ARv567es were detected in 67.53% and 29.87% of samples, respectively. Statistical analysis revealed a strong association of AR-V positivity with a history of second line hormonal therapies. CONCLUSIONS To our knowledge this is the first study to demonstrate that PAXgene preserved whole blood can be used to obtain clinically relevant information regarding the expression of 2 AR-Vs. These data on a castration resistant prostate cancer cohort support a role for AR-Vs in resistance to therapies targeting the AR ligand-binding domain.
Collapse
Affiliation(s)
- Xichun Liu
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Elisa Ledet
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana; Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Dongying Li
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ary Dotiwala
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana; Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Allie Steinberger
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana; Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Allison Feibus
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jianzhuo Li
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Yanfeng Qi
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jonathan Silberstein
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Benjamin Lee
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Yan Dong
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana; Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Haitao Zhang
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana; Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
39
|
Cao S, Zhan Y, Dong Y. Emerging data on androgen receptor splice variants in prostate cancer. Endocr Relat Cancer 2016; 23:T199-T210. [PMID: 27702752 PMCID: PMC5107136 DOI: 10.1530/erc-16-0298] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 12/13/2022]
Abstract
Androgen receptor splice variants are alternatively spliced variants of androgen receptor, which are C-terminally truncated and lack the canonical ligand-binding domain. Accumulating evidence has indicated a significant role of androgen receptor splice variants in mediating resistance of castration-resistant prostate cancer to current therapies and in predicting therapeutic responses. As such, there is an urgent need to target androgen receptor splicing variants for more effective treatment of castration-resistant prostate cancer. Identification of precise and critical targeting points to deactivate androgen receptor splicing variants relies on a deep understanding of how they are generated and the mechanisms of their action. In this review, we will focus on the emerging data on their generation, clinical significance and mechanisms of action as well as the therapeutic influence of these findings.
Collapse
Affiliation(s)
- Subing Cao
- College of Life SciencesJilin University, Changchun, Jilin, China
- Department of Structural and Cellular BiologyTulane University School of Medicine, Tulane Cancer Center, New Orleans, Louisiana, USA
| | - Yang Zhan
- College of Life SciencesJilin University, Changchun, Jilin, China
- Department of Structural and Cellular BiologyTulane University School of Medicine, Tulane Cancer Center, New Orleans, Louisiana, USA
| | - Yan Dong
- College of Life SciencesJilin University, Changchun, Jilin, China
- Department of Structural and Cellular BiologyTulane University School of Medicine, Tulane Cancer Center, New Orleans, Louisiana, USA
| |
Collapse
|
40
|
Antonarakis ES, Armstrong AJ, Dehm SM, Luo J. Androgen receptor variant-driven prostate cancer: clinical implications and therapeutic targeting. Prostate Cancer Prostatic Dis 2016; 19:231-41. [PMID: 27184811 PMCID: PMC5493501 DOI: 10.1038/pcan.2016.17] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/13/2022]
Abstract
While there are myriad mechanisms of primary and acquired resistance to conventional and next-generation hormonal therapies in prostate cancer, the potential role of androgen receptor splice variants (AR-Vs) has recently gained momentum. AR-Vs are abnormally truncated isoforms of the androgen receptor (AR) protein that lack the COOH-terminal domain but retain the NH2-terminal domain and DNA-binding domain and are thus constitutively active even in the absence of ligands. Although multiple preclinical studies have previously implicated AR-Vs in the development of castration resistance as well as resistance to abiraterone and enzalutamide, recent technological advances have made it possible to reliably detect and quantify AR-Vs from human clinical tumor specimens including blood samples. Initial clinical studies have now shown that certain AR-Vs, in particular AR-V7, may be associated with resistance to abiraterone and enzalutamide but not taxane chemotherapies when detected in circulating tumor cells. Efforts are now underway to clinically validate AR-V7 as a relevant treatment-selection biomarker in the context of other key genomic aberrations in men with metastatic castration-resistant prostate cancer. Additional efforts are underway to therapeutically target both AR and AR-Vs either directly or indirectly. Whether AR-Vs represent drivers of castration-resistant prostate cancer, or whether they are simply passenger events associated with aggressive disease or clonal heterogeneity, will ultimately be answered only through these types of clinical trials.
Collapse
MESH Headings
- Alternative Splicing
- Androgen Receptor Antagonists/therapeutic use
- Androgens/metabolism
- Animals
- Antineoplastic Agents, Hormonal/therapeutic use
- Biomarkers, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Clinical Trials as Topic
- Drug Evaluation, Preclinical
- Drug Resistance, Neoplasm/drug effects
- Epithelium/metabolism
- Epithelium/pathology
- Gene Expression Regulation, Neoplastic
- Genetic Variation
- Humans
- Male
- Molecular Targeted Therapy
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Protein Binding
- Protein Interaction Domains and Motifs
- Protein Multimerization
- Receptors, Androgen/chemistry
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Research
- Signal Transduction/drug effects
- Transcription, Genetic
- Treatment Outcome
Collapse
Affiliation(s)
- ES Antonarakis
- Departments of Oncology and Urology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - AJ Armstrong
- Departments of Medicine, Surgery, and Pharmacology and Cancer Biology, Divisions of Medical Oncology and Urology, Duke Cancer Institute, Durham, NC, USA
| | - SM Dehm
- Masonic Cancer Center and Departments of Laboratory Medicine and Pathology and Urology, University of Minnesota, Minneapolis, MN, USA
| | - J Luo
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
41
|
Abstract
Prostate cancer (PCa) is one of the most lethal cancers in western countries. Androgen receptor (AR) signaling pathway plays a key role in PCa progression. Despite the initial effectiveness of androgen deprivation therapy (ADT)for treatment of patients with advanced PCa, most of them will develop resistance to ADT and progress to metastatic castration resistant prostate cancer (mCRPC). Constitutively transcriptional activated AR splice variants (AR-Vs) have emerged as critical players in the development and progression of mCRPC. Among AR-Vs identified to date, AR-V7 (a.k.a. AR3) is one of the most abundant and frequently found in both PCa cell lines and in human prostate tissues. Most of functional studies have been focused on AR-V7/AR3 and revealed its role in regulation of survival, growth, differentiation and migration in prostate cells. In this review, we will summarize our current understanding of regulation of expression and activity of AR-Vs in mCRPC.
Collapse
Affiliation(s)
- Jin Xu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yun Qiu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
42
|
Ware KE, Somarelli JA, Schaeffer D, Li J, Zhang T, Park S, Patierno SR, Freedman J, Foo WC, Garcia MA, Armstrong AJ. Snail promotes resistance to enzalutamide through regulation of androgen receptor activity in prostate cancer. Oncotarget 2016; 7:50507-50521. [PMID: 27409172 PMCID: PMC5226599 DOI: 10.18632/oncotarget.10476] [Citation(s) in RCA: 35] [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: 04/29/2016] [Accepted: 06/20/2016] [Indexed: 11/25/2022] Open
Abstract
Treatment with androgen-targeted therapies can induce upregulation of epithelial plasticity pathways. Epithelial plasticity is known to be important for metastatic dissemination and therapeutic resistance. The goal of this study is to elucidate the functional consequence of induced epithelial plasticity on AR regulation during disease progression to identify factors important for treatment-resistant and metastatic prostate cancer. We pinpoint the epithelial plasticity transcription factor, Snail, at the nexus of enzalutamide resistance and prostate cancer metastasis both in preclinical models of prostate cancer and in patients. In patients, Snail expression is associated with Gleason 9-10 high-risk disease and is strongly overexpressed in metastases as compared to localized prostate cancer. Snail expression is also elevated in enzalutamide-resistant prostate cancer cells compared to enzalutamide-sensitive cells, and downregulation of Snail re-sensitizes enzalutamide-resistant cells to enzalutamide. While activation of Snail increases migration and invasion, it is also capable of promoting enzalutamide resistance in enzalutamide-sensitive cells. This Snail-mediated enzalutamide resistance is a consequence of increased full-length AR and AR-V7 expression and nuclear localization. Downregulation of either full-length AR or AR-V7 re-sensitizes cells to enzalutamide in the presence of Snail, thus connecting Snail-induced enzalutamide resistance directly to AR biology. Finally, we demonstrate that Snail is capable of mediating-resistance through AR even in the absence of AR-V7. These findings imply that increased Snail expression during progression to metastatic disease may prime cells for resistance to AR-targeted therapies by promoting AR activity in prostate cancer.
Collapse
Affiliation(s)
- Kathryn E. Ware
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Jason A. Somarelli
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Daneen Schaeffer
- Department of Oncology, Translational Research, Janssen Research and Development, Spring House, PA, USA
| | - Jing Li
- Department of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tian Zhang
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Sally Park
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Steven R. Patierno
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Jennifer Freedman
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Wen-Chi Foo
- Department of Pathology, Duke University, Durham, NC, USA
| | - Mariano A. Garcia
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Andrew J. Armstrong
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Genitourinary Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| |
Collapse
|
43
|
Austin DC, Strand DW, Love HL, Franco OE, Grabowska MM, Miller NL, Hameed O, Clark PE, Matusik RJ, Jin RJ, Hayward SW. NF-κB and androgen receptor variant 7 induce expression of SRD5A isoforms and confer 5ARI resistance. Prostate 2016; 76:1004-18. [PMID: 27197599 PMCID: PMC4912960 DOI: 10.1002/pros.23195] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 04/18/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is treated with 5α-reductase inhibitors (5ARI). These drugs inhibit the conversion of testosterone to dihydrotestosterone resulting in apoptosis and prostate shrinkage. Most patients initially respond to 5ARIs; however, failure is common especially in inflamed prostates, and often results in surgery. This communication examines a link between activation of NF-κB and increased expression of SRD5A2 as a potential mechanism by which patients fail 5ARI therapy. METHODS Tissue was collected from "Surgical" patients, treated specifically for lower urinary tract symptoms secondary to advanced BPH; and, cancer free transition zone from "Incidental" patients treated for low grade, localized peripheral zone prostate cancer. Clinical, molecular and histopathological profiles were analyzed. Human prostatic stromal and epithelial cell lines were genetically modified to regulate NF-κB activity, androgen receptor (AR) full length (AR-FL), and AR variant 7 (AR-V7) expression. RESULTS SRD5A2 is upregulated in advanced BPH. SRD5A2 was significantly associated with prostate volume determined by Transrectal Ultrasound (TRUS), and with more severe lower urinary tract symptoms (LUTS) determined by American Urological Association Symptom Score (AUASS). Synthesis of androgens was seen in cells in which NF-κB was activated. AR-FL and AR-V7 expression increased SRD5A2 expression while forced activation of NF-κB increased all three SRD5A isoforms. Knockdown of SRD5A2 in the epithelial cells resulted in significant reduction in proliferation, AR target gene expression, and response to testosterone (T). In tissue recombinants, canonical NF-κB activation in prostatic epithelium elevated all three SRD5A isoforms and resulted in in vivo growth under castrated conditions. CONCLUSION Increased BPH severity in patients correlates with SRD5A2 expression. We demonstrate that NF-κB and AR-V7 upregulate SRD5A expression providing a mechanism to explain failure of 5ARI therapy in BPH patients. Prostate 76:1004-1018, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David C. Austin
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas W. Strand
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Harold L. Love
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Omar E. Franco
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Magdalena M. Grabowska
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nicole L. Miller
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Omar Hameed
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Peter E. Clark
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J. Matusik
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ren J. Jin
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Simon W. Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| |
Collapse
|
44
|
Austin DC, Strand DW, Love HL, Franco OE, Jang A, Grabowska MM, Miller NL, Hameed O, Clark PE, Fowke JH, Matusik RJ, Jin RJ, Hayward SW. NF-κB and androgen receptor variant expression correlate with human BPH progression. Prostate 2016; 76:491-511. [PMID: 26709083 PMCID: PMC4763342 DOI: 10.1002/pros.23140] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is a common, chronic progressive disease. Inflammation is associated with prostatic enlargement and resistance to 5α-reductase inhibitor (5ARI) therapy. Activation of the nuclear factor-kappa B (NF-κB) pathway is linked to both inflammation and ligand-independent prostate cancer progression. METHODS NF-κB activation and androgen receptor variant (AR-V) expression were quantified in transition zone tissue samples from patients with a wide range of AUASS from incidental BPH in patients treated for low grade, localized peripheral zone prostate cancer to advanced disease requiring surgical intervention. To further investigate these pathways, human prostatic stromal and epithelial cell lines were transduced with constitutively active or kinase dead forms of IKK2 to regulate canonical NF-κB activity. The effects on AR full length (AR-FL) and androgen-independent AR-V expression as well as cellular growth and differentiation were assessed. RESULTS Canonical NF-κB signaling was found to be upregulated in late versus early stage BPH, and to be strongly associated with non-insulin dependent diabetes mellitus. Elevated expression of AR-variant 7 (AR-V7), but not other AR variants, was found in advanced BPH samples. Expression of AR-V7 significantly correlated with the patient AUASS and TRUS volume. Forced activation of canonical NF-κB in human prostatic epithelial and stromal cells resulted in elevated expression of both AR-FL and AR-V7, with concomitant ligand-independent activation of AR reporters. Activation of NF-κB and over expression of AR-V7 in human prostatic epithelial cells maintained cell viability in the face of 5ARI treatment. CONCLUSION Activation of NF-κB and AR-V7 in the prostate is associated with increased disease severity. AR-V7 expression is inducible in human prostate cells by forced activation of NF-κB resulting in resistance to 5ARI treatment, suggesting a potential mechanism by which patients may become resistant to 5ARI therapy.
Collapse
Affiliation(s)
- David C Austin
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas W Strand
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Harold L Love
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Omar E Franco
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Alex Jang
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Magdalena M Grabowska
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nicole L Miller
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Omar Hameed
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Peter E Clark
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jay H Fowke
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J Matusik
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ren J Jin
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Simon W Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| |
Collapse
|
45
|
Olokpa E, Bolden A, Stewart LV. The Androgen Receptor Regulates PPARγ Expression and Activity in Human Prostate Cancer Cells. J Cell Physiol 2016; 231:2664-72. [PMID: 26945682 PMCID: PMC5132088 DOI: 10.1002/jcp.25368] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/02/2016] [Indexed: 01/22/2023]
Abstract
The peroxisome proliferator activated receptor gamma (PPARγ) is a ligand-activated transcription factor that regulates growth and differentiation within normal prostate and prostate cancers. However the factors that control PPARγ within the prostate cancers have not been characterized. The goal of this study was to examine whether the androgen receptor (AR) regulates PPARγ expression and function within human prostate cancer cells. qRT-PCR and Western blot analyses revealed nanomolar concentrations of the AR agonist dihydrotestosterone (DHT) decrease PPARγ mRNA and protein within the castration-resistant, AR-positive C4-2 and VCaP human prostate cancer cell lines. The AR antagonists bicalutamide and enzalutamide blocked the ability of DHT to reduce PPARγ levels. In addition, siRNA mediated knockdown of AR increased PPARγ protein levels and ligand-induced PPARγ transcriptional activity within the C4-2 cell line. Furthermore, proteasome inhibitors that interfere with AR function increased the level of basal PPARγ and prevented the DHT-mediated suppression of PPARγ. These data suggest that AR normally functions to suppress PPARγ expression within AR-positive prostate cancer cells. To determine whether increases in AR protein would influence PPARγ expression and activity, we used lipofectamine-based transfections to overexpress AR within the AR-null PC-3 cells. The addition of AR to PC-3 cells did not significantly alter PPARγ protein levels. However, the ability of the PPARγ ligand rosiglitazone to induce activation of a PPARγ-driven luciferase reporter and induce expression of FABP4 was suppressed in AR-positive PC-3 cells. Together, these data indicate AR serves as a key modulator of PPARγ expression and function within prostate tumors. J. Cell. Physiol. 231: 2664-2672, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Emuejevoke Olokpa
- Department of Biochemistry and Cancer BiologyMeharry Medical CollegeNashvilleTennessee
| | - Adrienne Bolden
- Department of Biochemistry and Cancer BiologyMeharry Medical CollegeNashvilleTennessee
| | - LaMonica V. Stewart
- Department of Biochemistry and Cancer BiologyMeharry Medical CollegeNashvilleTennessee
| |
Collapse
|
46
|
Qin X, Liu M, Wang X. New insights into the androgen biotransformation in prostate cancer: A regulatory network among androgen, androgen receptors and UGTs. Pharmacol Res 2016; 106:114-122. [PMID: 26926093 DOI: 10.1016/j.phrs.2016.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 01/15/2023]
Abstract
Androgen, as one kind of steroid hormones, is pivotal in the hormone-sensitive cancer, such as prostate cancer (PCa). The synthesis, elimination, and bioavailability of androgen in prostate cells have been proved to be a main cause of the carcinogenesis, maintenance and deterioration of PCa. This review illustrates the outlines of androgen biotransformation, and further discusses the different enzymes, especially UDP-glucuronyltransferases (UGTs) embedded in both benign and malignant prostate cells, which catalyze the reactions. Although many inhibitors of the enzymes responsible for the synthesis of androgens have been developed into drugs to fight against PCa, the elimination procedures metabolized by the UGTs are less emphasized. Thus the regulatory network among androgen, androgen receptors (AR) and UGTs is carefully reviewed in this article, indicating the determinant effects of UGTs on prostatic androgens and the regulation of AR. Finally, the hypothesis is also put forward that the regulators of UGTs may be developed to accelerate the androgen elimination and benefit PCa therapy.
Collapse
Affiliation(s)
- Xuan Qin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, TX, USA
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
47
|
Mikhaylenko DS, Efremov GD, Sivkov AV, Zaletaev DV. Hormone resistance and neuroendocrine differentiation due to accumulation of genetic lesions during clonal evolution of prostate cancer. Mol Biol 2016. [DOI: 10.1134/s0026893315060187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
48
|
Lam HM, Ho SM, Chen J, Medvedovic M, Tam NNC. Bisphenol A Disrupts HNF4α-Regulated Gene Networks Linking to Prostate Preneoplasia and Immune Disruption in Noble Rats. Endocrinology 2016; 157:207-19. [PMID: 26496021 PMCID: PMC4701889 DOI: 10.1210/en.2015-1363] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Exposure of humans to bisphenol A (BPA) is widespread and continuous. The effects of protracted exposure to BPA on the adult prostate have not been studied. We subjected Noble rats to 32 weeks of BPA (low or high dose) or 17β-estradiol (E2) in conjunction with T replenishment. T treatment alone or untreated groups were used as controls. Circulating T levels were maintained within the physiological range in all treatment groups, whereas the levels of free BPA were elevated in the groups treated with T+low BPA (1.06 ± 0.05 ng/mL, P < .05) and T+high BPA (10.37 ± 0.43 ng/mL, P < .01) when compared with those in both controls (0.1 ± 0.05 ng/mL). Prostatic hyperplasia, low-grade prostatic intraepithelial neoplasia (PIN), and marked infiltration of CD4+ and CD8+ T cells into the PIN epithelium (P < .05) were observed in the lateral prostates (LPs) of T+low/high BPA-treated rats. In contrast, only hyperplasia and high-grade PIN, but no aberrant immune responses, were found in the T+E2-treated LPs. Genome-wide transcriptome analysis in LPs identified differential changes between T+BPA vs T+E2 treatment. Expression of multiple genes in the regulatory network controlled by hepatocyte nuclear factor 4α was perturbed by the T+BPA but not by the T+E2 exposure. Collectively these findings suggest that the adult rat prostate, under a physiologically relevant T environment, is susceptible to BPA-induced transcriptomic reprogramming, immune disruption, and aberrant growth dysregulation in a manner distinct from those caused by E2. They are more relevant to our recent report of higher urinary levels BPA found in patients with prostate cancer than those with benign disease.
Collapse
Affiliation(s)
- Hung-Ming Lam
- Department of Environmental Health (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Center for Environmental Genetics (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Cincinnati Cancer Center (S.-M.H., M.M., N.N.C.T.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; and Cincinnati Veteran Affairs Hospital Medical Center (S.-M.H.), Cincinnati, Ohio 45220
| | - Shuk-Mei Ho
- Department of Environmental Health (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Center for Environmental Genetics (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Cincinnati Cancer Center (S.-M.H., M.M., N.N.C.T.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; and Cincinnati Veteran Affairs Hospital Medical Center (S.-M.H.), Cincinnati, Ohio 45220
| | - Jing Chen
- Department of Environmental Health (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Center for Environmental Genetics (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Cincinnati Cancer Center (S.-M.H., M.M., N.N.C.T.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; and Cincinnati Veteran Affairs Hospital Medical Center (S.-M.H.), Cincinnati, Ohio 45220
| | - Mario Medvedovic
- Department of Environmental Health (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Center for Environmental Genetics (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Cincinnati Cancer Center (S.-M.H., M.M., N.N.C.T.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; and Cincinnati Veteran Affairs Hospital Medical Center (S.-M.H.), Cincinnati, Ohio 45220
| | - Neville Ngai Chung Tam
- Department of Environmental Health (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Center for Environmental Genetics (H.-M.L., S.-M.H., J.C., M.M., N.N.C.T.), Cincinnati Cancer Center (S.-M.H., M.M., N.N.C.T.), University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; and Cincinnati Veteran Affairs Hospital Medical Center (S.-M.H.), Cincinnati, Ohio 45220
| |
Collapse
|
49
|
Ciccarese C, Santoni M, Brunelli M, Buti S, Modena A, Nabissi M, Artibani W, Martignoni G, Montironi R, Tortora G, Massari F. AR-V7 and prostate cancer: The watershed for treatment selection? Cancer Treat Rev 2015; 43:27-35. [PMID: 26827690 DOI: 10.1016/j.ctrv.2015.12.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 12/06/2015] [Accepted: 12/11/2015] [Indexed: 12/20/2022]
Abstract
The androgen receptor (AR) plays a key role in progression to metastatic castration-resistant prostate cancer (mCRPC). Despite the recent progress in targeting persistent AR activity with the next-generation hormonal therapies (abiraterone acetate and enzalutamide), resistance to these agents limits therapeutic efficacy for many patients. Several explanations for response and/or resistance to abiraterone acetate and enzalutamide are emerging, but growing interest is focusing on importance of AR splice variants (AR-Vs) and in particular of AR-V7. Increasing evidences highlight the concept that variant expression could be used as a potential predictive biomarker and a therapeutic target in advanced prostate cancer. Therefore, understanding the mechanisms of treatment resistance or sensitivity can help to achieve a more effective management of mCRPC, increasing clinical outcomes and representing a promising and engaging area of prostate cancer research.
Collapse
Affiliation(s)
- Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Matteo Santoni
- Medical Oncology, AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, A.O.U.I., University of Verona, Verona, Italy
| | | | - Alessandra Modena
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Massimo Nabissi
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy
| | - Walter Artibani
- Urologic Clinic, Department of Oncological and Surgical Sciences, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Pathology and Diagnostic, A.O.U.I., University of Verona, Verona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Francesco Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy.
| |
Collapse
|
50
|
Watson PA, Arora VK, Sawyers CL. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer 2015; 15:701-11. [PMID: 26563462 PMCID: PMC4771416 DOI: 10.1038/nrc4016] [Citation(s) in RCA: 1070] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the past 10 years, preclinical studies implicating sustained androgen receptor (AR) signalling as the primary driver of castration-resistant prostate cancer (CRPC) have led to the development of novel agents targeting the AR pathway that are now in widespread clinical use. These drugs prolong the survival of patients with late-stage prostate cancer but are not curative. In this Review, we highlight emerging mechanisms of acquired resistance to these contemporary therapies, which fall into the three broad categories of restored AR signalling, AR bypass signalling and complete AR independence. This diverse range of resistance mechanisms presents new challenges for long-term disease control, which may be addressable through early use of combination therapies guided by recent insights from genomic landscape studies of CRPC.
Collapse
Affiliation(s)
- Philip A Watson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Vivek K Arora
- Division of Medical Oncology, Washington University School of Medicine, St Louis, Missouri 63130, USA
| | - Charles L Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| |
Collapse
|