1
|
Walker L, Duncan R, Adamson B, Kendall H, Brittain N, Luzzi S, Jones D, Chaytor L, Peel S, Crafter C, O’Neill DJ, Gaughan L. Defining Splicing Factor Requirements for Androgen Receptor Variant Synthesis in Advanced Prostate Cancer. Mol Cancer Res 2024; 22:1128-1142. [PMID: 39348093 PMCID: PMC11612623 DOI: 10.1158/1541-7786.mcr-23-0958] [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] [Received: 03/15/2024] [Revised: 08/02/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Resistance to androgen receptor (AR)-targeted therapies represents a major challenge in prostate cancer. A key mechanism of treatment resistance in patients who progress to castration-resistant prostate cancer (CRPC) is the generation of alternatively spliced AR variants (AR-V). Unlike full-length AR isoforms, AR-Vs are constitutively active and refractory to current receptor-targeting agents and hence drive tumor progression. Identifying regulators of AR-V synthesis may therefore provide new therapeutic opportunities in combination with conventional AR-targeting agents. Our understanding of AR transcript splicing, and the factors that control the synthesis of AR-Vs, remains limited. Although candidate-based approaches have identified a small number of AR-V splicing regulators, an unbiased analysis of splicing factors important for AR-V generation is required to fill an important knowledge gap and furnish the field with novel and tractable targets for prostate cancer treatment. To that end, we conducted a bespoke CRISPR screen to profile splicing factor requirements for AR-V synthesis. MFAP1 and CWC22 were shown to be required for the generation of AR-V mRNA transcripts, and their depletion resulted in reduced AR-V protein abundance and cell proliferation in several CRPC models. Global transcriptomic analysis of MFAP1-depleted cells revealed both AR-dependent and -independent transcriptional impacts, including genes associated with DNA damage response. As such, MFAP1 downregulation sensitized prostate cancer cells to ionizing radiation, suggesting that therapeutically targeting AR-V splicing could provide novel cellular vulnerabilities which can be exploited in CRPC. Implications: We have utilized a CRISPR screening approach to identify key regulators of pathogenic AR splicing in prostate cancer.
Collapse
Affiliation(s)
- Laura Walker
- Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Ruaridh Duncan
- Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Beth Adamson
- Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | - Hannah Kendall
- Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| | | | - Sara Luzzi
- Newcastle University Biosciences Institute, International Centre for Life, Newcastle upon Tyne, United Kingdom
| | | | - Lewis Chaytor
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Samantha Peel
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Claire Crafter
- Early Oncology, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Daniel J. O’Neill
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Luke Gaughan
- Newcastle University Centre for Cancer, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
2
|
Lu S, Lamba M, Wang J, Dong Z. Targeting proliferating cell nuclear antigen enhances ionizing radiation-induced cytotoxicity in prostate cancer cells. Prostate 2024; 84:1456-1467. [PMID: 39219052 DOI: 10.1002/pros.24786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/30/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Proliferating cell nuclear antigen (PCNA) is essential for DNA replication and repair, cell growth, and survival. PCNA also enhances androgen receptor (AR) signaling in prostate cancer (PC) cells. We identified a PCNA interaction protein (PIP) box at the N-terminal domain of AR and developed a small peptide PCNA inhibitor R9-AR-PIP containing AR PIP-box. We also identified a series of small molecule PCNA inhibitors (PCNA-Is) that bind directly to PCNA and interrupt PCNA functions. The present study investigated the effects of the PCNA inhibitors on the sensitivity of PC cells to X-ray radiation. METHODS The effects of targeting PCNA on radio sensitivity of PC cells were investigated in four lines of castration-resistant PC (CRPC) cells with different AR expression statuses. The cells were treated with the PCNA inhibitors and X-ray radiation alone or in combination. The effects of the treatment on expression of AR target genes, DNA damage response, DNA damage, homologous recombination repair (HRR), and cytotoxicity were evaluated. RESULTS We found that the androgen response element (ARE) occupancy of the DNA damage response gene PARP1 by AR is significantly attenuated by PCNA-I1S or R9-AR-PIP combined with X-ray radiation, while X-ray radiation alone does not enhance the ARE occupancy. PCNA-I1S or R9-AR-PIP alone significantly inhibits occupancy of the AR-occupied regions (AROR) in PRKDC and XRCC2 genes. R9-AR-PIP and PCNA-I1S inhibit expression of AR-Vs target gene cyclin A2 and show the additive effects with radiation in AR-positive CRPC cells. Targeting PCNA by PCNA-I1S and R9-AR-PIP downregulates expression of DNA damage response genes EXO1, Rad54L, Rad51, and/or PARP1 and shows the additive effects with radiation as compared with their respective controls in AR-positive CRPC LNCaP-AI, 22Rv1, and R1-D567 cells, but not in AR-negative PC-3 cells. R9-AR-PIP and PCNA-I1S elevate the levels of phospho-DNA-PKcs(S2056) and γH2AX, indicating DNA damage in response to radiation in AR-positive cells. The HRR is significantly attenuated by PCNA inhibitors PCNA-I1S, R9-AR-PIP, and T2AA in all four CRPC cells examined, and inhibited by Enzalutamide (Enz) only in 22RV1 cells. The cytotoxicity induced by X-ray radiation in androgen-dependent LNCaP cells is enhanced by Enz and a lower concentration of R9-AR-PIP in the colony formation assay. R9-AR-PIP at higher concentration reduces the colony formation and has an additive effect with X-ray radiation in all AR expressing cells, regardless of AR-FL and AR-Vs, but does not significantly alter the colony formation in AR-negative PC-3 cells. PCNA-I1S attenuates colony formation and has an additive effect with ionizing radiation in all four CRPC cells, regardless of AR expression status. CONCLUSION These data provide a strong rationale for the therapy studies using PCNA-I1S or R9-AR-PIP in combination with X-ray radiation against CRPC tumors in preclinical models.
Collapse
Affiliation(s)
- Shan Lu
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michael Lamba
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jiang Wang
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Zhongyun Dong
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
3
|
Tabibian M, Moghaddam FS, Motevaseli E, Ghafouri-Fard S. Targeting mRNA-coding genes in prostate cancer using CRISPR/Cas9 technology with a special focus on androgen receptor signaling. Cell Commun Signal 2024; 22:504. [PMID: 39420406 PMCID: PMC11484332 DOI: 10.1186/s12964-024-01833-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Prostate cancer is among prevalent cancers in men. Numerous strategies have been proposed to intervene with the important prostate cancer-related signaling pathways. Among the most promising strategies is CRISPR/Cas9 strategy. This strategy has been used to modify expression of a number of genes in prostate cancer cells. AIMS This review summarizes the most recent progresses in the application of CRISPR/Cas9 strategy in modification of prostate cancer-related phenotypes with an especial focus on pathways related to androgen receptor signaling. CONCLUSION CRISPR/Cas9 technology has successfully targeted several genes in the prostate cancer cells. Moreover, the efficiency of this technique in reducing tumor burden has been tested in animal models of prostate cancer. Most of targeted genes have been related with the androgen receptor signaling. Targeted modulation of these genes have affected growth of castration-resistant prostate cancer. PI3K/AKT/mTOR signaling and immune response-related genes have been other targets that have been successfully modulated by CRISPR/Cas9 technology in prostate cancer. Based on the rapid translation of this technology into the clinical application, it is anticipated that novel treatments based on this technique change the outcome of this malignancy in future.
Collapse
Affiliation(s)
- Mobina Tabibian
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and Biotechnologies, Shahid Beheshti University, Tehran, Iran
| | | | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Kostos L, Tran B, Azad AA. Combination of PARP Inhibitors and Androgen Receptor Pathway Inhibitors in Metastatic Castration-Resistant Prostate Cancer. Drugs 2024; 84:1093-1109. [PMID: 39060912 PMCID: PMC11438617 DOI: 10.1007/s40265-024-02071-y] [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: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Despite recent advances in the treatment of metastatic prostate cancer, progression to a castration-resistant state remains inevitable for most and prognosis is limited. Genetic testing for homologous recombination repair pathway alterations is recommended for all patients with advanced prostate cancer given that a mutation is present in up to 25% of cases. Poly(ADP-ribose) polymerase (PARPis) are now approved for use in patients with metastatic castration-resistant prostate cancer who have progressed on an androgen receptor pathway inhibitor (ARPI) and harbour a germline or somatic homologous recombination repair mutation. Preclinical data support a synergistic effect with an ARPI and PARPi, and various ARPI-PARPi combinations have therefore been explored in phase III clinical trials. Despite heterogeneous findings, a clear hierarchy of benefit is evident, with patients harbouring a BRCA mutation deriving the greatest magnitude of benefit, followed by any homologous recombination repair mutation. The benefit in homologous recombination repair-proficient cohort is less clear, and questions remain about whether ARPI-PARPi combination therapy should be offered to patients without a homologous recombination repair mutation. With ARPIs now considered standard-of-care for metastatic hormone-sensitive prostate cancer, ARPI-PARPi combination therapy is currently being explored earlier in the treatment paradigm. The purpose of this review is to discuss the rationale behind ARPI-PARPi combination therapy, summarise the results of key clinical trials, and discuss clinical considerations and future perspectives.
Collapse
Affiliation(s)
- Louise Kostos
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Ben Tran
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Arun A Azad
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Garofoli M, Maiorano BA, Bruno G, Giordano G, Di Tullio P, Maselli FM, Landriscina M, Conteduca V. Androgen receptor, PARP signaling, and tumor microenvironment: the 'perfect triad' in prostate cancer? Ther Adv Med Oncol 2024; 16:17588359241258443. [PMID: 38887656 PMCID: PMC11181896 DOI: 10.1177/17588359241258443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Aberrations in the homologous recombination repair (HRR) pathway in prostate cancer (PCa) provide a unique opportunity to develop therapeutic strategies that take advantage of the reduced tumor ability to repair DNA damage. Poly-ADP-ribose polymerase (PARP) inhibitors (PARPi) have been shown to prolong the survival of PCa patients with HRR defects, particularly in those with Breast Cancer type 1 susceptibility protein/Breast Cancer type 2 susceptibility protein alterations. To expand the benefit of PARPi to patients without detectable HRR alterations, multiple preclinical and clinical studies are addressing potential synergies between PARPi and androgen receptor signaling inhibitors, and these strategies are also being evaluated in combination with other drugs such as immune checkpoint inhibitors. However, the effectiveness of these combining therapies could be hindered by multiple mechanisms of resistance, including also the role played by the immunosuppressive tumor microenvironment. In this review, we summarize the use of PARPi in PCa and the potential synergies with different molecular pathways. However, numerous unanswered questions remain, including the identification of the patient population that could benefit most from PARPi, determining whether to use PARPi as monotherapy or in combination, and finding the optimal timing of PARPi, expanding the use of genomic tests, and optimizing combination therapies.
Collapse
Affiliation(s)
- Marianna Garofoli
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | | | - Giuseppina Bruno
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Piergiorgio Di Tullio
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Felicia Maria Maselli
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Matteo Landriscina
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Viale Pinto, 1, Foggia 71122, Italy
| | - Vincenza Conteduca
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Viale Pinto, 1, Foggia 71122, Italy
| |
Collapse
|
6
|
Ma T, Jin L, Bai S, Liu Z, Wang S, Shen B, Cho Y, Cao S, Sun MJS, Fazli L, Zhang D, Wedderburn C, Zhang DY, Mugon G, Ungerleider N, Baddoo M, Zhang K, Schiavone LH, Burkhardt BR, Fan J, You Z, Flemington EK, Dong X, Dong Y. Loss of feedback regulation between FAM3B and androgen receptor driving prostate cancer progression. J Natl Cancer Inst 2024; 116:421-433. [PMID: 37847647 PMCID: PMC10919334 DOI: 10.1093/jnci/djad215] [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] [Received: 05/24/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Although the fusion of the transmembrane serine protease 2 gene (TMPRSS2) with the erythroblast transformation-specific-related gene (ERG), or TMPRSS2-ERG, occurs frequently in prostate cancer, its impact on clinical outcomes remains controversial. Roughly half of TMPRSS2-ERG fusions occur through intrachromosomal deletion of interstitial genes and the remainder via insertional chromosomal rearrangements. Because prostate cancers with deletion-derived TMPRSS2-ERG fusions are more aggressive than those with insertional fusions, we investigated the impact of interstitial gene loss on prostate cancer progression. METHODS We conducted an unbiased analysis of transcriptome data from large collections of prostate cancer samples and employed diverse in vitro and in vivo models combined with genetic approaches to characterize the interstitial gene loss that imposes the most important impact on clinical outcome. RESULTS This analysis identified FAM3B as the top-ranked interstitial gene whose loss is associated with a poor prognosis. The association between FAM3B loss and poor clinical outcome extended to fusion-negative prostate cancers where FAM3B downregulation occurred through epigenetic imprinting. Importantly, FAM3B loss drives disease progression in prostate cancer. FAM3B acts as an intermediator of a self-governing androgen receptor feedback loop. Specifically, androgen receptor upregulates FAM3B expression by binding to an intronic enhancer to induce an enhancer RNA and facilitate enhancer-promoter looping. FAM3B, in turn, attenuates androgen receptor signaling. CONCLUSION Loss of FAM3B in prostate cancer, whether through the TMPRSS2-ERG translocation or epigenetic imprinting, causes an exit from this autoregulatory loop to unleash androgen receptor activity and prostate cancer progression. These findings establish FAM3B loss as a new driver of prostate cancer progression and support the utility of FAM3B loss as a biomarker to better define aggressive prostate cancer.
Collapse
Affiliation(s)
- Tianfang Ma
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Lianjin Jin
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Shanshan Bai
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Zhan Liu
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Shuo Wang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Beibei Shen
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yeyoung Cho
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Subing Cao
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Meijuan J S Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ladan Fazli
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - David Zhang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Duke University, Durham, NC, USA
| | - Chiyaro Wedderburn
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Derek Y Zhang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- University of Southern California, Los Angeles, CA, USA
| | - Gavisha Mugon
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nathan Ungerleider
- Department of Pathology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Melody Baddoo
- Department of Pathology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Kun Zhang
- Department of Computer Science, Bioinformatics Facility of Xavier RCMI Center of Cancer Research, Xavier University of Louisiana, New Orleans, LA, USA
| | | | - Brant R Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Jia Fan
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - Erik K Flemington
- Department of Pathology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
| | - Xuesen Dong
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Yan Dong
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| |
Collapse
|
7
|
Garnham R, Geh D, Nelson R, Ramon-Gil E, Wilson L, Schmidt EN, Walker L, Adamson B, Buskin A, Hepburn AC, Hodgson K, Kendall H, Frame FM, Maitland N, Coffey K, Strand DW, Robson CN, Elliott DJ, Heer R, Macauley M, Munkley J, Gaughan L, Leslie J, Scott E. ST3 beta-galactoside alpha-2,3-sialyltransferase 1 (ST3Gal1) synthesis of Siglec ligands mediates anti-tumour immunity in prostate cancer. Commun Biol 2024; 7:276. [PMID: 38448753 PMCID: PMC10918101 DOI: 10.1038/s42003-024-05924-0] [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: 04/11/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Immune checkpoint blockade has yet to produce robust anti-cancer responses for prostate cancer. Sialyltransferases have been shown across several solid tumours, including breast, melanoma, colorectal and prostate to promote immune suppression by synthesising sialoglycans, which act as ligands for Siglec receptors. We report that ST3 beta-galactoside alpha-2,3-sialyltransferase 1 (ST3Gal1) levels negatively correlate with androgen signalling in prostate tumours. We demonstrate that ST3Gal1 plays an important role in modulating tumour immune evasion through the synthesises of sialoglycans with the capacity to engage the Siglec-7 and Siglec-9 immunoreceptors preventing immune clearance of cancer cells. Here, we provide evidence of the expression of Siglec-7/9 ligands and their respective immunoreceptors in prostate tumours. These interactions can be modulated by enzalutamide and may maintain immune suppression in enzalutamide treated tumours. We conclude that the activity of ST3Gal1 is critical to prostate cancer anti-tumour immunity and provide rationale for the use of glyco-immune checkpoint targeting therapies in advanced prostate cancer.
Collapse
Affiliation(s)
- Rebecca Garnham
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Daniel Geh
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Ryan Nelson
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Erik Ramon-Gil
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Laura Wilson
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Laura Walker
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Beth Adamson
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Adriana Buskin
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Anastasia C Hepburn
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Kirsty Hodgson
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Hannah Kendall
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Fiona M Frame
- Cancer Research Unit, Department of Biology, University of York, Heslington, North Yorkshire, YO10 5DD, UK
| | - Norman Maitland
- Cancer Research Unit, Department of Biology, University of York, Heslington, North Yorkshire, YO10 5DD, UK
| | - Kelly Coffey
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Douglas W Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Craig N Robson
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - David J Elliott
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Rakesh Heer
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Matthew Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Jennifer Munkley
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Luke Gaughan
- Newcastle University, Centre for Cancer, Newcastle University Translational and Clinical Research Institute, Newcastle, NE1 3BZ, UK
| | - Jack Leslie
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK
| | - Emma Scott
- Newcastle University, Centre for Cancer, Newcastle University Biosciences Institute, Newcastle, NE1 3BZ, UK.
| |
Collapse
|
8
|
Fizazi K, Azad AA, Matsubara N, Carles J, Fay AP, De Giorgi U, Joung JY, Fong PCC, Voog E, Jones RJ, Shore ND, Dunshee C, Zschäbitz S, Oldenburg J, Ye D, Lin X, Healy CG, Di Santo N, Laird AD, Zohren F, Agarwal N. First-line talazoparib with enzalutamide in HRR-deficient metastatic castration-resistant prostate cancer: the phase 3 TALAPRO-2 trial. Nat Med 2024; 30:257-264. [PMID: 38049622 PMCID: PMC10803259 DOI: 10.1038/s41591-023-02704-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023]
Abstract
Preclinical evidence has suggested an interplay between the androgen receptor, which largely drives the growth of prostate cancer cells, and poly(ADP-ribose) polymerase. This association provides a rationale for their co-inhibition for the treatment of metastatic castration-resistant prostate cancer (mCRPC), an area of unmet medical need. The phase 3 TALAPRO-2 study investigated combining the poly(ADP-ribose) polymerase inhibitor talazoparib with enzalutamide versus enzalutamide alone as first-line treatment of mCRPC. Patients were prospectively assessed for tumor alterations in DNA damage response genes involved in homologous recombination repair (HRR). Two cohorts were enrolled sequentially: an all-comers cohort that was enrolled first (cohort 1; N = 805 (169 were HRR-deficient)), followed by an HRR-deficient-only cohort (cohort 2; N = 230). We present results from the alpha-controlled primary analysis for the combined HRR-deficient population (N = 399). Patients were randomized in a 1:1 ratio to talazoparib or placebo, plus enzalutamide. The primary endpoint, radiographic progression-free survival, was met (median not reached at the time of the analysis for the talazoparib group versus 13.8 months for the placebo group; hazard ratio, 0.45; 95% confidence interval, 0.33 to 0.61; P < 0.0001). Data for overall survival, a key secondary endpoint, are immature but favor talazoparib (hazard ratio, 0.69; 95% confidence interval, 0.46 to 1.03; P = 0.07). Common adverse events in the talazoparib group were anemia, fatigue and neutropenia. Combining talazoparib with enzalutamide significantly improved radiographic progression-free survival in patients with mCRPC harboring HRR gene alterations, supporting talazoparib plus enzalutamide as a potential first-line treatment for these patients. ClinicalTrials.gov Identifier: NCT03395197 .
Collapse
Affiliation(s)
- Karim Fizazi
- Institut Gustave Roussy, University of Paris-Saclay, Villejuif, France.
| | - Arun A Azad
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Joan Carles
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Andre P Fay
- PUCRS School of Medicine, Porto Alegre, Brazil
| | - Ugo De Giorgi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) Dino Amadori, Meldola, Italy
| | | | - Peter C C Fong
- Auckland City Hospital, Auckland, New Zealand
- University of Auckland, Auckland, New Zealand
| | - Eric Voog
- Clinique Victor Hugo Centre Jean Bernard, Le Mans, France
| | - Robert J Jones
- School of Cancer Sciences, University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | | | - Stefanie Zschäbitz
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Jan Oldenburg
- Akershus University Hospital (Ahus), Lørenskog, Norway
| | - Dingwei Ye
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xun Lin
- Pfizer Inc., La Jolla, CA, USA
| | | | | | | | | | - Neeraj Agarwal
- Huntsman Cancer Institute (NCI-CCC), University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
9
|
Stracker TH, Osagie OI, Escorcia FE, Citrin DE. Exploiting the DNA Damage Response for Prostate Cancer Therapy. Cancers (Basel) 2023; 16:83. [PMID: 38201511 PMCID: PMC10777950 DOI: 10.3390/cancers16010083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Prostate cancers that progress despite androgen deprivation develop into castration-resistant prostate cancer, a fatal disease with few treatment options. In this review, we discuss the current understanding of prostate cancer subtypes and alterations in the DNA damage response (DDR) that can predispose to the development of prostate cancer and affect its progression. We identify barriers to conventional treatments, such as radiotherapy, and discuss the development of new therapies, many of which target the DDR or take advantage of recurring genetic alterations in the DDR. We place this in the context of advances in understanding the genetic variation and immune landscape of CRPC that could help guide their use in future treatment strategies. Finally, we discuss several new and emerging agents that may advance the treatment of lethal disease, highlighting selected clinical trials.
Collapse
Affiliation(s)
- Travis H. Stracker
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (O.I.O.); (F.E.E.); (D.E.C.)
| | - Oloruntoba I. Osagie
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (O.I.O.); (F.E.E.); (D.E.C.)
| | - Freddy E. Escorcia
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (O.I.O.); (F.E.E.); (D.E.C.)
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Deborah E. Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (O.I.O.); (F.E.E.); (D.E.C.)
| |
Collapse
|
10
|
Calabrese M, Saporita I, Turco F, Gillessen S, Castro E, Vogl UM, Di Stefano RF, Carfì FM, Poletto S, Farinea G, Tucci M, Buttigliero C. Synthetic Lethality by Co-Inhibition of Androgen Receptor and Polyadenosine Diphosphate-Ribose in Metastatic Prostate Cancer. Int J Mol Sci 2023; 25:78. [PMID: 38203248 PMCID: PMC10779404 DOI: 10.3390/ijms25010078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Androgen receptor pathway inhibitors (ARPI) and polyadenosine diphosphate-ribose inhibitors (PARPi) are part of the standard of care in patients with metastatic castration-resistant prostate cancer (mCRPC). There is biological evidence that the association of ARPI and PARPi could have a synergistic effect; therefore, several ongoing clinical trials are investigating the efficacy of this combination with preliminary results that are not perfectly concordant in identifying patients who can obtain the most benefit from this therapeutic option. The purpose of this review is to describe the PARPi mechanisms of action and to analyze the biological mechanisms behind the interplay between the androgen receptor and the PARPi system to better understand the rationale of the ARPI + PARPi combinations. Furthermore, we will summarize the preliminary results of the ongoing studies on these combinations, trying to understand in which patients to apply. Finally, we will discuss the clinical implications of this combination and its possible future perspectives.
Collapse
Affiliation(s)
- Mariangela Calabrese
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Isabella Saporita
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Fabio Turco
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
- Ente Ospedaliero Cantonale—Istituto Oncologico della Svizzera Italiana, 6500 Bellinzona, Switzerland; (S.G.); (U.M.V.)
| | - Silke Gillessen
- Ente Ospedaliero Cantonale—Istituto Oncologico della Svizzera Italiana, 6500 Bellinzona, Switzerland; (S.G.); (U.M.V.)
- Department of Medical Oncology, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Elena Castro
- Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Ursula Maria Vogl
- Ente Ospedaliero Cantonale—Istituto Oncologico della Svizzera Italiana, 6500 Bellinzona, Switzerland; (S.G.); (U.M.V.)
| | - Rosario Francesco Di Stefano
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Federica Maria Carfì
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Stefano Poletto
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Giovanni Farinea
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| | - Marcello Tucci
- Department of Medical Oncology, Cardinal Massaia Hospital, 14100 Asti, Italy;
| | - Consuelo Buttigliero
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy; (M.C.); (I.S.); (F.T.); (R.F.D.S.); (F.M.C.); (S.P.); (G.F.)
| |
Collapse
|
11
|
Adamson B, Brittain N, Walker L, Duncan R, Luzzi S, Rescigno P, Smith G, McGill S, Burchmore RJ, Willmore E, Hickson I, Robson CN, Bogdan D, Jimenez-Vacas JM, Paschalis A, Welti J, Yuan W, McCracken SR, Heer R, Sharp A, de Bono JS, Gaughan L. The catalytic subunit of DNA-PK regulates transcription and splicing of AR in advanced prostate cancer. J Clin Invest 2023; 133:e169200. [PMID: 37751307 PMCID: PMC10645393 DOI: 10.1172/jci169200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023] Open
Abstract
Aberrant androgen receptor (AR) signaling drives prostate cancer (PC), and it is a key therapeutic target. Although initially effective, the generation of alternatively spliced AR variants (AR-Vs) compromises efficacy of treatments. In contrast to full-length AR (AR-FL), AR-Vs constitutively activate androgenic signaling and are refractory to the current repertoire of AR-targeting therapies, which together drive disease progression. There is an unmet clinical need, therefore, to develop more durable PC therapies that can attenuate AR-V function. Exploiting the requirement of coregulatory proteins for AR-V function has the capacity to furnish tractable routes for attenuating persistent oncogenic AR signaling in advanced PC. DNA-PKcs regulates AR-FL transcriptional activity and is upregulated in both early and advanced PC. We hypothesized that DNA-PKcs is critical for AR-V function. Using a proximity biotinylation approach, we demonstrated that the DNA-PK holoenzyme is part of the AR-V7 interactome and is a key regulator of AR-V-mediated transcription and cell growth in models of advanced PC. Crucially, we provide evidence that DNA-PKcs controls global splicing and, via RBMX, regulates the maturation of AR-V and AR-FL transcripts. Ultimately, our data indicate that targeting DNA-PKcs attenuates AR-V signaling and provide evidence that DNA-PKcs blockade is an effective therapeutic option in advanced AR-V-positive patients with PC.
Collapse
Affiliation(s)
- Beth Adamson
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Nicholas Brittain
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Laura Walker
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Ruaridh Duncan
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Sara Luzzi
- Newcastle University Biosciences Institute, International Centre for Life, Newcastle Upon Tyne, United Kingdom
| | - Pasquale Rescigno
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Graham Smith
- Newcastle University Bioinformatics Support Unit, Medical School, Newcastle Upon Tyne, United Kingdom
| | - Suzanne McGill
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Richard J.S. Burchmore
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Elaine Willmore
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Ian Hickson
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Craig N. Robson
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Denisa Bogdan
- The Institute for Cancer Research, London, United Kingdom
| | | | - Alec Paschalis
- The Institute for Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Jonathan Welti
- The Institute for Cancer Research, London, United Kingdom
| | - Wei Yuan
- The Institute for Cancer Research, London, United Kingdom
| | - Stuart R. McCracken
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| | - Rakesh Heer
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
- Division of Surgery, Imperial College London, London, United Kingdom
| | - Adam Sharp
- The Institute for Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Johann S. de Bono
- The Institute for Cancer Research, London, United Kingdom
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Luke Gaughan
- Newcastle University Centre for Cancer, Paul O’Gorman Building, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
12
|
Imamura J, Ganguly S, Muskara A, Liao RS, Nguyen JK, Weight C, Wee CE, Gupta S, Mian OY. Lineage plasticity and treatment resistance in prostate cancer: the intersection of genetics, epigenetics, and evolution. Front Endocrinol (Lausanne) 2023; 14:1191311. [PMID: 37455903 PMCID: PMC10349394 DOI: 10.3389/fendo.2023.1191311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Androgen deprivation therapy is a cornerstone of treatment for advanced prostate cancer, and the development of castrate-resistant prostate cancer (CRPC) is the primary cause of prostate cancer-related mortality. While CRPC typically develops through a gain in androgen receptor (AR) signaling, a subset of CRPC will lose reliance on the AR. This process involves genetic, epigenetic, and hormonal changes that promote cellular plasticity, leading to AR-indifferent disease, with neuroendocrine prostate cancer (NEPC) being the quintessential example. NEPC is enriched following treatment with second-generation anti-androgens and exhibits resistance to endocrine therapy. Loss of RB1, TP53, and PTEN expression and MYCN and AURKA amplification appear to be key drivers for NEPC differentiation. Epigenetic modifications also play an important role in the transition to a neuroendocrine phenotype. DNA methylation of specific gene promoters can regulate lineage commitment and differentiation. Histone methylation can suppress AR expression and promote neuroendocrine-specific gene expression. Emerging data suggest that EZH2 is a key regulator of this epigenetic rewiring. Several mechanisms drive AR-dependent castration resistance, notably AR splice variant expression, expression of the adrenal-permissive 3βHSD1 allele, and glucocorticoid receptor expression. Aberrant epigenetic regulation also promotes radioresistance by altering the expression of DNA repair- and cell cycle-related genes. Novel therapies are currently being developed to target these diverse genetic, epigenetic, and hormonal mechanisms promoting lineage plasticity-driven NEPC.
Collapse
Affiliation(s)
- Jarrell Imamura
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Shinjini Ganguly
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Andrew Muskara
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Ross S. Liao
- Glickman Urologic Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jane K. Nguyen
- Glickman Urologic Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Christopher Weight
- Glickman Urologic Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Christopher E. Wee
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Omar Y. Mian
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
13
|
Constantin TA, Varela-Carver A, Greenland KK, de Almeida GS, Olden E, Penfold L, Ang S, Ormrod A, Leach DA, Lai CF, Ainscow EK, Bahl AK, Carling D, Fuchter MJ, Ali S, Bevan CL. The CDK7 inhibitor CT7001 (Samuraciclib) targets proliferation pathways to inhibit advanced prostate cancer. Br J Cancer 2023; 128:2326-2337. [PMID: 37076563 PMCID: PMC10241923 DOI: 10.1038/s41416-023-02252-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Current strategies to inhibit androgen receptor (AR) are circumvented in castration-resistant prostate cancer (CRPC). Cyclin-dependent kinase 7 (CDK7) promotes AR signalling, in addition to established roles in cell cycle and global transcription, providing a rationale for its therapeutic targeting in CRPC. METHODS The antitumour activity of CT7001, an orally bioavailable CDK7 inhibitor, was investigated across CRPC models in vitro and in xenograft models in vivo. Cell-based assays and transcriptomic analyses of treated xenografts were employed to investigate the mechanisms driving CT7001 activity, alone and in combination with the antiandrogen enzalutamide. RESULTS CT7001 selectively engages with CDK7 in prostate cancer cells, causing inhibition of proliferation and cell cycle arrest. Activation of p53, induction of apoptosis, and suppression of transcription mediated by full-length and constitutively active AR splice variants contribute to antitumour efficacy in vitro. Oral administration of CT7001 represses growth of CRPC xenografts and significantly augments growth inhibition achieved by enzalutamide. Transcriptome analyses of treated xenografts indicate cell cycle and AR inhibition as the mode of action of CT7001 in vivo. CONCLUSIONS This study supports CDK7 inhibition as a strategy to target deregulated cell proliferation and demonstrates CT7001 is a promising CRPC therapeutic, alone or in combination with AR-targeting compounds.
Collapse
Affiliation(s)
- Theodora A Constantin
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Anabel Varela-Carver
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Kyle K Greenland
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Gilberto Serrano de Almeida
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Ellen Olden
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Lucy Penfold
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Simon Ang
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Alice Ormrod
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Damien A Leach
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Chun-Fui Lai
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Edward K Ainscow
- Carrick Therapeutics, Nova UCD, Bellfield Innovation Park, Dublin, 4, Ireland
| | - Ash K Bahl
- Carrick Therapeutics, Nova UCD, Bellfield Innovation Park, Dublin, 4, Ireland
| | - David Carling
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Matthew J Fuchter
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, London, UK
| | - Simak Ali
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK.
| |
Collapse
|
14
|
Liu J, Zhao Y, He D, Jones KM, Tang S, Allison DB, Zhang Y, Chen J, Zhang Q, Wang X, Li C, Wang C, Li L, Liu X. A kinome-wide CRISPR screen identifies CK1α as a target to overcome enzalutamide resistance of prostate cancer. Cell Rep Med 2023; 4:101015. [PMID: 37075701 PMCID: PMC10140619 DOI: 10.1016/j.xcrm.2023.101015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 01/13/2023] [Accepted: 03/21/2023] [Indexed: 04/21/2023]
Abstract
Enzalutamide (ENZA), a second-generation androgen receptor antagonist, has significantly increased progression-free and overall survival of patients with metastatic prostate cancer (PCa). However, resistance remains a prominent obstacle in treatment. Utilizing a kinome-wide CRISPR-Cas9 knockout screen, we identified casein kinase 1α (CK1α) as a therapeutic target to overcome ENZA resistance. Depletion or pharmacologic inhibition of CK1α enhanced ENZA efficacy in ENZA-resistant cells and patient-derived xenografts. Mechanistically, CK1α phosphorylates the serine residue S1270 and modulates the protein abundance of ataxia telangiectasia mutated (ATM), a primary initiator of DNA double-strand break (DSB)-response signaling, which is compromised in ENZA-resistant cells and patients. Inhibition of CK1α stabilizes ATM, resulting in the restoration of DSB signaling, and thus increases ENZA-induced cell death and growth arrest. Our study details a therapeutic approach for ENZA-resistant PCa and characterizes a particular perspective for the function of CK1α in the regulation of DNA-damage response.
Collapse
Affiliation(s)
- Jinghui Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Yue Zhao
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Katelyn M Jones
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Shan Tang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Derek B Allison
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Yanquan Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Qiongsi Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Xinyi Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Chaohao Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Lang Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
15
|
Soave C, Ducker C, Islam N, Kim S, Yurgelevic S, Nicely NI, Pardy L, Huang Y, Shaw PE, Auner G, Dickson A, Ratnam M. The Small Molecule Antagonist KCI807 Disrupts Association of the Amino-Terminal Domain of the Androgen Receptor with ELK1 by Modulating the Adjacent DNA Binding Domain. Mol Pharmacol 2023; 103:211-220. [PMID: 36720643 PMCID: PMC11033959 DOI: 10.1124/molpharm.122.000589] [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] [Received: 07/12/2022] [Revised: 12/11/2022] [Accepted: 12/27/2022] [Indexed: 02/02/2023] Open
Abstract
The androgen receptor (AR) is a crucial coactivator of ELK1 for prostate cancer (PCa) growth, associating with ELK1 through two peptide segments (358-457 and 514-557) within the amino-terminal domain (NTD) of AR. The small-molecule antagonist 5-hydroxy-2-(3-hydroxyphenyl)chromen-4-one (KCI807) binds to AR, blocking ELK1 binding and inhibiting PCa growth. We investigated the mode of interaction of KCI807 with AR using systematic mutagenesis coupled with ELK1 coactivation assays, testing polypeptide binding and Raman spectroscopy. In full-length AR, deletion of neither ELK1 binding segment affected sensitivity of residual ELK1 coactivation to KCI807. Although the NTD is sufficient for association of AR with ELK1, interaction of the isolated NTD with ELK1 was insensitive to KCI807. In contrast, coactivation of ELK1 by the AR-V7 splice variant, comprising the NTD and the DNA binding domain (DBD), was sensitive to KCI807. Deletions and point mutations within DBD segment 558-595, adjacent to the NTD, interfered with coactivation of ELK1, and residual ELK1 coactivation by the mutants was insensitive to KCI807. In a glutathione S-transferase pull-down assay, KCI807 inhibited ELK1 binding to an AR polypeptide that included the two ELK1 binding segments and the DBD but did not affect ELK1 binding to a similar AR segment that lacked the sequence downstream of residue 566. Raman spectroscopy detected KCI807-induced conformational change in the DBD. The data point to a putative KCI807 binding pocket within the crystal structure of the DBD and indicate that either mutations or binding of KCI807 at this site will induce conformational changes that disrupt ELK1 binding to the NTD. SIGNIFICANCE STATEMENT: The small-molecule antagonist KCI807 disrupts association of the androgen receptor (AR) with ELK1, serving as a prototype for the development of small molecules for a novel type of therapeutic intervention in drug-resistant prostate cancer. This study provides basic information needed for rational KCI807-based drug design by identifying a putative binding pocket in the DNA binding domain of AR through which KCI807 modulates the amino-terminal domain to inhibit ELK1 binding.
Collapse
Affiliation(s)
- Claire Soave
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Charles Ducker
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Naeyma Islam
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Seongho Kim
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Sally Yurgelevic
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Nathan I Nicely
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Luke Pardy
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Yanfang Huang
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Peter E Shaw
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Gregory Auner
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Alex Dickson
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| | - Manohar Ratnam
- Department of Oncology (C.S., S.K., Y.H., L.P., M.R.) and Smart Sensors and Integrated Microsystems (SSIM) Program (S.Y., G.A.), Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan; Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University, East Lansing, Michigan (N.I. and A.D.); School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham, United Kingdom (C.D. and P.E.S.); and Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina (N.N.)
| |
Collapse
|
16
|
Choupani E, Mahmoudi Gomari M, Zanganeh S, Nasseri S, Haji-Allahverdipoor K, Rostami N, Hernandez Y, Najafi S, Saraygord-Afshari N, Hosseini A. Newly Developed Targeted Therapies Against the Androgen Receptor in Triple-Negative Breast Cancer: A Review. Pharmacol Rev 2023; 75:309-327. [PMID: 36781219 DOI: 10.1124/pharmrev.122.000665] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/26/2022] [Accepted: 10/31/2022] [Indexed: 12/15/2022] Open
Abstract
Among different types of breast cancers (BC), triple-negative BC (TNBC) amounts to 15% to 20% of breast malignancies. Three principal characteristics of TNBC cells are (i) extreme aggressiveness, (ii) absence of hormones, and (iii) growth factor receptors. Due to the lack or poor expression of the estrogen receptor, human epidermal growth factor receptor 2, and progesterone receptor, TNBC is resistant to hormones and endocrine therapies. Consequently, chemotherapy is currently used as the primary approach against TNBC. Expression of androgen receptor (AR) in carcinoma cells has been observed in a subset of patients with TNBC; therefore, inhibiting androgen signaling pathways holds promise for TNBC targeting. The new AR inhibitors have opened up new therapy possibilities for BC patients carrying AR-positive TNBC cells. Our group provides a comprehensive review of the structure and function of the AR and clinical evidence for targeting the cell's nuclear receptor in TNBC. We updated AR agonists, inhibitors, and antagonists. We also presented a new era of genetic manipulating CRISPR/Cas9 and nanotechnology as state-of-the-art approaches against AR to promote the efficiency of targeted therapy in TNBC. SIGNIFICANCE STATEMENT: The lack of effective treatment for triple-negative breast cancer is a health challenge. The main disadvantages of existing treatments are their side effects, due to their nonspecific targeting. Molecular targeting of cellular receptors, such as androgen receptors, increased expression in malignant tissues, significantly improving the survival rate of breast cancer patients.
Collapse
Affiliation(s)
- Edris Choupani
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Mohammad Mahmoudi Gomari
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Saeed Zanganeh
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Sherko Nasseri
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Kaveh Haji-Allahverdipoor
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Neda Rostami
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Yaeren Hernandez
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Safa Najafi
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Neda Saraygord-Afshari
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| | - Arshad Hosseini
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran (E.C., M.M.G., N.S.-A., A.H.); Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran (S.Z.); Department of Molecular Medicine, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran (S.N., K.H.-a.); Department of Chemical Engineering, Faculty of Engineering, Arak University, Iran (N.R.); Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona (Y.H.); and Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran (S.N.)
| |
Collapse
|
17
|
Dhital B, Santasusagna S, Kirthika P, Xu M, Li P, Carceles-Cordon M, Soni RK, Li Z, Hendrickson RC, Schiewer MJ, Kelly WK, Sternberg CN, Luo J, Lujambio A, Cordon-Cardo C, Alvarez-Fernandez M, Malumbres M, Huang H, Ertel A, Domingo-Domenech J, Rodriguez-Bravo V. Harnessing transcriptionally driven chromosomal instability adaptation to target therapy-refractory lethal prostate cancer. Cell Rep Med 2023; 4:100937. [PMID: 36787737 PMCID: PMC9975292 DOI: 10.1016/j.xcrm.2023.100937] [Citation(s) in RCA: 11] [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: 08/12/2022] [Revised: 09/27/2022] [Accepted: 01/18/2023] [Indexed: 02/16/2023]
Abstract
Metastatic prostate cancer (PCa) inevitably acquires resistance to standard therapy preceding lethality. Here, we unveil a chromosomal instability (CIN) tolerance mechanism as a therapeutic vulnerability of therapy-refractory lethal PCa. Through genomic and transcriptomic analysis of patient datasets, we find that castration and chemotherapy-resistant tumors display the highest CIN and mitotic kinase levels. Functional genomics screening coupled with quantitative phosphoproteomics identify MASTL kinase as a survival vulnerability specific of chemotherapy-resistant PCa cells. Mechanistically, MASTL upregulation is driven by transcriptional rewiring mechanisms involving the non-canonical transcription factors androgen receptor splice variant 7 and E2F7 in a circuitry that restrains deleterious CIN and prevents cell death selectively in metastatic therapy-resistant PCa cells. Notably, MASTL pharmacological inhibition re-sensitizes tumors to standard therapy and improves survival of pre-clinical models. These results uncover a targetable mechanism promoting high CIN adaptation and survival of lethal PCa.
Collapse
Affiliation(s)
- Brittiny Dhital
- Biochemistry and Molecular Biology Department, Mayo Clinic, Rochester, MN 55905, USA; Urology Department, Mayo Clinic, Rochester, MN 55905, USA; Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Sandra Santasusagna
- Biochemistry and Molecular Biology Department, Mayo Clinic, Rochester, MN 55905, USA; Urology Department, Mayo Clinic, Rochester, MN 55905, USA
| | - Perumalraja Kirthika
- Biochemistry and Molecular Biology Department, Mayo Clinic, Rochester, MN 55905, USA; Urology Department, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael Xu
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Peiyao Li
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | | | - Rajesh K Soni
- Microchemistry and Proteomics Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zhuoning Li
- Microchemistry and Proteomics Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ronald C Hendrickson
- Microchemistry and Proteomics Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Matthew J Schiewer
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - William K Kelly
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Department of Medicine, Meyer Cancer Center, New York-Presbyterian Hospital, New York, NY 10021, USA
| | - Jun Luo
- Urology Department, Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amaia Lujambio
- Oncological Sciences Department, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carlos Cordon-Cardo
- Pathology Department, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Monica Alvarez-Fernandez
- Head & Neck Cancer Department, Institute de Investigación Sanitaria Principado de Asturias (ISPA), Institute Universitario de Oncología Principado de Asturias (IUOPA), 33011 Oviedo, Spain
| | - Marcos Malumbres
- Cell Division & Cancer Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; Cancer Cell Cycle group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Haojie Huang
- Biochemistry and Molecular Biology Department, Mayo Clinic, Rochester, MN 55905, USA; Urology Department, Mayo Clinic, Rochester, MN 55905, USA
| | - Adam Ertel
- Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Josep Domingo-Domenech
- Biochemistry and Molecular Biology Department, Mayo Clinic, Rochester, MN 55905, USA; Urology Department, Mayo Clinic, Rochester, MN 55905, USA.
| | - Veronica Rodriguez-Bravo
- Biochemistry and Molecular Biology Department, Mayo Clinic, Rochester, MN 55905, USA; Urology Department, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
18
|
Androgen receptor-dependent regulation of metabolism in high grade bladder cancer cells. Sci Rep 2023; 13:1762. [PMID: 36720985 PMCID: PMC9889754 DOI: 10.1038/s41598-023-28692-z] [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: 07/01/2022] [Accepted: 01/23/2023] [Indexed: 02/01/2023] Open
Abstract
The observed sex disparity in bladder cancer (BlCa) argues that androgen receptor (AR) signaling has a role in these malignancies. BlCas express full-length AR (FL-AR), constitutively active AR splice variants, including AR-v19, or both, and their depletion limits BlCa viability. However, the mechanistic basis of AR-dependence is unknown. Here, we depleted FL-AR, AR-v19, or all AR forms (T-AR), and performed RNA-seq studies to uncover that different AR forms govern distinct but partially overlapping transcriptional programs. Overlapping alterations include a decrease in mTOR and an increase of hypoxia regulated transcripts accompanied by a decline in oxygen consumption rate (OCR). Queries of BlCa databases revealed a significant negative correlation between AR expression and multiple hypoxia-associated transcripts arguing that this regulatory mechanism is a feature of high-grade malignancies. Our analysis of a 1600-compound library identified niclosamide as a strong ATPase inhibitor that reduces OCR in BlCa cells, decreased cell viability and induced apoptosis in a dose and time dependent manner. These results suggest that BlCa cells hijack AR signaling to enhance metabolic activity, promoting cell proliferation and survival; hence targeting this AR downstream vulnerability presents an attractive strategy to limit BlCa.
Collapse
|
19
|
Cathepsin K regulates the tumor growth and metastasis by IL-17/CTSK/EMT axis and mediates M2 macrophage polarization in castration-resistant prostate cancer. Cell Death Dis 2022; 13:813. [PMID: 36138018 PMCID: PMC9499936 DOI: 10.1038/s41419-022-05215-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 01/23/2023]
Abstract
A common stage of advanced prostate cancer is castration-resistant prostate cancer (CRPC), greater understanding of which is required in order to address and solve the clinically difficult challenge. Cathepsin K (CTSK) is a cysteine protease that usually has a strong activity of degrading extracellular matrix and is related to osteoclast-mediated bone destruction. However, the mechanism of CTSK-regulation in CRPC is still unclear to us. The current study aimed to analyze the expression of differentially expressed genes (DEGs) in patient samples (from localized PC and CRPC). Interestingly, we found that CTSK to be significantly up-regulated in CRPC. Through further signal pathway enrichment analysis, we found that the IL-17 signaling pathway to be highly correlated with CTSK. The oncogenic functions of CTSK and IL-17 in CRPC were proven by a series of in vivo and in vitro experiments. Possible downstream molecules of CTSK were investigated, which could serve as control elements to regulate the expression of EMT, thereby facilitating the metastasis and excessive proliferation of PC cells. Expression of CTSK was related to high concentration of M2 tumor-associated macrophages (TAMs) M2 in CRPC. A CTSK-mediated feedback circuit between TAMs and CRPC tissues was indicated in the process of transfer, proving the possibility of CTSK could be use as an available therapeutic target for CRPC.
Collapse
|
20
|
Tolkach Y, Kremer A, Lotz G, Schmid M, Mayr T, Förster S, Garbe S, Hosni S, Cronauer MV, Kocsmár I, Kocsmár É, Riesz P, Alajati A, Ritter M, Ellinger J, Ohlmann CH, Kristiansen G. Androgen Receptor Splice Variants Contribute to the Upregulation of DNA Repair in Prostate Cancer. Cancers (Basel) 2022; 14:4441. [PMID: 36139600 PMCID: PMC9496991 DOI: 10.3390/cancers14184441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Canonical androgen receptor (AR) signaling regulates a network of DNA repair genes in prostate cancer (PCA). Experimental and clinical evidence indicates that androgen deprivation not only suppresses DNA repair activity but is often synthetically lethal in combination with PARP inhibition. The present study aimed to elucidate the impact of AR splice variants (AR-Vs), occurring in advanced or late-stage PCA, on DNA repair machinery. METHODS Two hundred and seventy-three tissue samples were analyzed, including primary hormone-naïve PCA, primary metastases, hormone-sensitive PCA on androgen deprivation therapy (ADT) and castration refractory PCA (CRPC group). The transcript levels of the target genes were profiled using the nCounter platform. Experimental support for the findings was gained in AR/AR-V7-expressing LNCaP cells subjected to ionizing radiation. RESULTS AR-Vs were present in half of hormone-sensitive PCAs on androgen deprivation therapy (ADT) and two-thirds of CRPC samples. The presence of AR-Vs is highly correlated with increased activity in the AR pathway and DNA repair gene expression. In AR-V-expressing CRPC, the DNA repair score increased by 2.5-fold as compared to AR-V-negative samples. Enhanced DNA repair and the deregulation of DNA repair genes by AR-V7 supported the clinical data in a cell line model. CONCLUSIONS The expression of AR splice variants such as AR-V7 in PCA patients following ADT might be a reason for reduced or absent therapy effects in patients on additional PARP inhibition due to the modulation of DNA repair gene expression. Consequently, AR-Vs should be further studied as predictive biomarkers for therapy response in this setting.
Collapse
Affiliation(s)
- Yuri Tolkach
- Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany
| | - Anika Kremer
- Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Gábor Lotz
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Matthias Schmid
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, 53127 Bonn, Germany
| | - Thomas Mayr
- Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Sarah Förster
- Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Stephan Garbe
- Department of Radiation Oncology, University Hospital Bonn, 53127 Bonn, Germany
| | - Sana Hosni
- Clinic of Urology, University Hospital Bonn, 53127 Bonn, Germany
| | | | - Ildikó Kocsmár
- Department of Urology, Semmelweis University, 1085 Budapest, Hungary
| | - Éva Kocsmár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Riesz
- Department of Urology, Semmelweis University, 1085 Budapest, Hungary
| | - Abdullah Alajati
- Clinic of Urology, University Hospital Bonn, 53127 Bonn, Germany
| | - Manuel Ritter
- Clinic of Urology, University Hospital Bonn, 53127 Bonn, Germany
| | - Jörg Ellinger
- Clinic of Urology, University Hospital Bonn, 53127 Bonn, Germany
| | | | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
21
|
Luo H, Liu Y, Li Y, Zhang C, Yu B, Shao C. Androgen receptor splicing variant 7 (ARv7) promotes DNA damage response in prostate cancer cells. FASEB J 2022; 36:e22495. [PMID: 35947121 DOI: 10.1096/fj.202200190r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/13/2022] [Accepted: 08/01/2022] [Indexed: 11/11/2022]
Abstract
In the treatment of patients with locally advanced prostate cancer (PCa), androgen deprivation therapy (ADT) significantly enhances the efficacy of radiotherapy by weakening the DNA damage response (DDR) pathway. Recently, several studies have suggested that androgen receptor splicing variants (ARvs) may mediate a compensatory DDR pathway when canonical androgen receptor (AR) signaling is inhibited, thus contributing to the resistance of some patients to this combinational treatment. However, the specific roles of certain ARvs as well as the detailed mechanism of how ARvs regulate the DDR are not well understood. Here, we demonstrated that AR splicing variant 7 (ARv7), which is the most abundant form of ARvs, significantly promotes the DDR of PCa cells under severe DNA damage independent of its parental AR by using the ionizing radiation (IR) and doxorubicin (Dox)-treated cell models. Mechanistically, ARv7 is sufficient to upregulate both the homologous recombination (HR) and the nonhomologous end joining (NHEJ) pathways by forming a positive regulatory loop with poly ADP-ribose polymerase 1 (PARP1). Moreover, the presence of ARv7 impairs the synergistic effect between AR antagonists and poly ADP-ribose polymerase (PARP) inhibitor, which has been recently shown to be a promising future treatment strategy for metastatic castration resistant prostate cancer (mCRPC). Combined, our data indicate that constitutively active ARv7 not only contributes to radioresistance after ADT, but may also serve as a potential predictive biomarker for assessing the efficacy of novel PARP inhibitor-based therapy in PCa.
Collapse
Affiliation(s)
- Haoge Luo
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yanan Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yang Li
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chaoke Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Bingbing Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chen Shao
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
22
|
Nevedomskaya E, Haendler B. From Omics to Multi-Omics Approaches for In-Depth Analysis of the Molecular Mechanisms of Prostate Cancer. Int J Mol Sci 2022; 23:6281. [PMID: 35682963 PMCID: PMC9181488 DOI: 10.3390/ijms23116281] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/24/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer arises following alterations at different cellular levels, including genetic and epigenetic modifications, transcription and translation dysregulation, as well as metabolic variations. High-throughput omics technologies that allow one to identify and quantify processes involved in these changes are now available and have been instrumental in generating a wealth of steadily increasing data from patient tumors, liquid biopsies, and from tumor models. Extensive investigation and integration of these data have led to new biological insights into the origin and development of multiple cancer types and helped to unravel the molecular networks underlying this complex pathology. The comprehensive and quantitative analysis of a molecule class in a biological sample is named omics and large-scale omics studies addressing different prostate cancer stages have been performed in recent years. Prostate tumors represent the second leading cancer type and a prevalent cause of cancer death in men worldwide. It is a very heterogenous disease so that evaluating inter- and intra-tumor differences will be essential for a precise insight into disease development and plasticity, but also for the development of personalized therapies. There is ample evidence for the key role of the androgen receptor, a steroid hormone-activated transcription factor, in driving early and late stages of the disease, and this led to the development and approval of drugs addressing diverse targets along this pathway. Early genomic and transcriptomic studies have allowed one to determine the genes involved in prostate cancer and regulated by androgen signaling or other tumor-relevant signaling pathways. More recently, they have been supplemented by epigenomic, cistromic, proteomic and metabolomic analyses, thus, increasing our knowledge on the intricate mechanisms involved, the various levels of regulation and their interplay. The comprehensive investigation of these omics approaches and their integration into multi-omics analyses have led to a much deeper understanding of the molecular pathways involved in prostate cancer progression, and in response and resistance to therapies. This brings the hope that novel vulnerabilities will be identified, that existing therapies will be more beneficial by targeting the patient population likely to respond best, and that bespoke treatments with increased efficacy will be available soon.
Collapse
Affiliation(s)
| | - Bernard Haendler
- Research and Early Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany;
| |
Collapse
|
23
|
Ganapathy K, Ngo C, Andl T, Coppola D, Park J, Chakrabarti R. Anti-cancer function of microRNA-30e is mediated by negative regulation of HELLPAR, a noncoding macroRNA, and genes involved in ubiquitination and cell cycle progression in prostate cancer. Mol Oncol 2022; 16:2936-2958. [PMID: 35612714 PMCID: PMC9394257 DOI: 10.1002/1878-0261.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/08/2022] [Accepted: 03/17/2022] [Indexed: 11/07/2022] Open
Abstract
Prostate cancer (PCa) progression relies on androgen receptor (AR) function, making AR a top candidate for PCa therapy. However, development of drug resistance is common, which eventually leads to development of castration‐resistant PCa. This warrants a better understanding of the pathophysiology of PCa that facilitates the aberrant activation of key signaling pathways including AR. MicroRNAs (miRNAs) function as regulators of cancer progression as they modulate various cellular processes. Here, we demonstrate a multidimensional function of miR‐30e through the regulation of genes involved in various signaling pathways. We noted loss of miR‐30e expression in prostate tumors, which, when restored, led to cell cycle arrest, induction of apoptosis, improved drug sensitivity of PCa cells and reduced tumor progression in xenograft models. We show that experimental upregulation of miR‐30e reduces expression of mRNAs including AR, FBXO45, SRSF7 and MYBL2 and a novel long noncoding RNA (lncRNA) HELLPAR, which are involved in cell cycle, apoptosis and ubiquitination, and the effects could be rescued by inhibition of miR‐30e expression. RNA immunoprecipitation analysis confirmed direct interactions between miR‐30e and its RNA targets. We noted a newly identified reciprocal relationship between miR‐30e and HELLPAR, as inhibition of HELLPAR improved stabilization of miR‐30e. Transcriptome profiling and quantitative real‐time PCR (qRT‐PCR) validation of miR‐30e‐expressing PCa cells showed differential expression of genes involved in cell cycle progression, apoptosis and ubiquitination, which supports our in vitro study. This study demonstrates an integrated function of miR‐30e on dysregulation of miRNA/lncRNA/mRNA axes that may have diagnostic and therapeutic significance in aggressive PCa.
Collapse
Affiliation(s)
- Kavya Ganapathy
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Christopher Ngo
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Domenico Coppola
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida, USA.,Florida Digestive Health Specialists, Bradenton, Florida, USA
| | - Jong Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Ratna Chakrabarti
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
24
|
Cistrome and transcriptome analysis identifies unique androgen receptor (AR) and AR-V7 splice variant chromatin binding and transcriptional activities. Sci Rep 2022; 12:5351. [PMID: 35354884 PMCID: PMC8969163 DOI: 10.1038/s41598-022-09371-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/14/2022] [Indexed: 12/13/2022] Open
Abstract
The constitutively active androgen receptor (AR) splice variant, AR-V7, plays an important role in resistance to androgen deprivation therapy in castration resistant prostate cancer (CRPC). Studies seeking to determine whether AR-V7 is a partial mimic of the AR, or also has unique activities, and whether the AR-V7 cistrome contains unique binding sites have yielded conflicting results. One limitation in many studies has been the low level of AR variant compared to AR. Here, LNCaP and VCaP cell lines in which AR-V7 expression can be induced to match the level of AR, were used to compare the activities of AR and AR-V7. The two AR isoforms shared many targets, but overall had distinct transcriptomes. Optimal induction of novel targets sometimes required more receptor isoform than classical targets such as PSA. The isoforms displayed remarkably different cistromes with numerous differential binding sites. Some of the unique AR-V7 sites were located proximal to the transcription start sites (TSS). A de novo binding motif similar to a half ARE was identified in many AR-V7 preferential sites and, in contrast to conventional half ARE sites that bind AR-V7, FOXA1 was not enriched at these sites. This supports the concept that the AR isoforms have unique actions with the potential to serve as biomarkers or novel therapeutic targets.
Collapse
|
25
|
Agarwal N, Azad A, Shore ND, Carles J, Fay AP, Dunshee C, Karsh LI, Paccagnella ML, Santo ND, Elmeliegy M, Lin X, Czibere A, Fizazi K. Talazoparib plus enzalutamide in metastatic castration-resistant prostate cancer: TALAPRO-2 Phase III study design. Future Oncol 2022; 18:425-436. [PMID: 35080190 DOI: 10.2217/fon-2021-0811] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PARP inhibitors in combination with androgen receptor-targeted therapy have demonstrated potential in the treatment of metastatic castration-resistant prostate cancer (mCRPC). Here, we describe the design and rationale of the multinational, Phase III, two-part TALAPRO-2 study comparing talazoparib plus enzalutamide versus placebo plus enzalutamide as a first-line treatment for patients with mCRPC with or without DNA damage response (DDR) alterations. This study has two co-primary end points: radiographic progression-free survival (rPFS) by blinded independent clinical review in all-comers (Cohort 1) and in patients with DDR alterations (Cohort 2). TALAPRO-2 will demonstrate whether talazoparib plus enzalutamide can significantly improve the efficacy of enzalutamide in terms of rPFS in both molecularly unselected and DDR-deficient patients with mCRPC (NCT03395197). Clinical Trial Registration: NCT03395197 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Neeraj Agarwal
- Huntsman Cancer Institute (NCI-CCC), University of Utah, Salt Lake City, UT 84112, USA
| | - Arun Azad
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Neal D Shore
- Department of Urology, Carolina Urologic Research Center, Myrtle Beach, SC 29572, USA
| | - Joan Carles
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona 08035, Spain
| | - Andre P Fay
- PUCRS School of Medicine Grupo Oncoclínicas, Porto Alegre 90610-000, Brazil
| | - Curtis Dunshee
- Urological Associates of Southern Arizona, Tucson, AZ 85741, USA
| | | | | | - Nicola Di Santo
- Pfizer Inc., Global Product Development, Durham, NC 27707, USA
| | | | - Xun Lin
- Pfizer Inc., Global Product Development, La Jolla, 92121 CA, USA
| | | | - Karim Fizazi
- Department of Cancer Medicine, Institut Gustave Roussy, University of Paris Saclay, Villejuif 94800, France
| |
Collapse
|
26
|
AR Structural Variants and Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:195-211. [DOI: 10.1007/978-3-031-11836-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Özgün F, Kaya Z, Morova T, Geverts B, Abraham TE, Houtsmuller AB, van Royen ME, Lack NA. DNA binding alters ARv7 dimer interactions. J Cell Sci 2021; 134:jcs258332. [PMID: 34318896 DOI: 10.1242/jcs.258332] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/02/2021] [Indexed: 11/20/2022] Open
Abstract
Androgen receptor (AR) splice variants are proposed to be a potential driver of lethal castration-resistant prostate cancer. AR splice variant 7 (ARv7) is the most commonly observed isoform and strongly correlates with resistance to second-generation anti-androgens. Despite this clinical evidence, the interplay between ARv7 and the highly expressed full-length AR (ARfl) remains unclear. In this work, we show that ARfl/ARv7 heterodimers readily form in the nucleus via an intermolecular N/C interaction that brings the four termini of the proteins in close proximity. Combining fluorescence resonance energy transfer and fluorescence recovery after photobleaching, we demonstrate that these heterodimers undergo conformational changes following DNA binding, indicating dynamic nuclear receptor interaction. Although transcriptionally active, ARv7 can only form short-term interactions with DNA at highly accessible high-occupancy ARfl binding sites. Dimerization with ARfl does not affect ARv7 binding dynamics, suggesting that DNA binding occupancy is determined by the individual protein monomers and not the homodimer or heterodimer complex. Overall, these biophysical studies reveal detailed properties of ARv7 dynamics as both a homodimer or heterodimer with ARfl.
Collapse
Affiliation(s)
- Fatma Özgün
- School of Medicine, Koç University, Istanbul 34450, Turkey
| | - Zeynep Kaya
- School of Medicine, Koç University, Istanbul 34450, Turkey
| | - Tunç Morova
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Bart Geverts
- Erasmus Optical Imaging Centre, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Tsion E Abraham
- Erasmus Optical Imaging Centre, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Adriaan B Houtsmuller
- Erasmus Optical Imaging Centre, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
- Department of Pathology, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Martin E van Royen
- Erasmus Optical Imaging Centre, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
- Department of Pathology, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Nathan A Lack
- School of Medicine, Koç University, Istanbul 34450, Turkey
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| |
Collapse
|
28
|
McFarland TR, Kessel A, Swami U, Agarwal N. Development of PARP inhibitor combinations for castration resistant prostate cancer unselected for homologous recombination repair mutations. Am J Transl Res 2021; 13:7427-7439. [PMID: 34377227 PMCID: PMC8340210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/10/2021] [Indexed: 06/13/2023]
Abstract
Genetic instability is a hallmark of cancer and, with the introduction of poly (ADP-ribose) polymerase (PARP) inhibitors, is a targetable feature of many tumors. Currently, two PARP inhibitors, olaparib and rucaparib, have received approval as monotherapy by the Food and Drug Administration for the treatment of men with castration resistant prostate cancer with selected mutations involving the homologous recombination (HR) pathway. However, it is currently debated whether an HR mutation is a prerequisite for response or if patients with HR-proficient mCRPC may also benefit from their use when combined with other targeted or immunotherapeutic agents. Several large phase III trials of PARP inhibitors with novel androgen axis inhibitors in groups of unselected patients are underway. Additionally, there are several early phase trials combining PARP inhibitors with radioligands or immunecheckpoint inhibitors. Here we discuss the currently ongoing or recently concluded trials of PARP inhibitor based combinatorial therapies in unselected patients with mCRPC, the rationale behind these trials, and how these may impact the treatment paradigm in men with mCRPC.
Collapse
Affiliation(s)
- Taylor Ryan McFarland
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah Salt Lake, UT, USA
| | - Adam Kessel
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah Salt Lake, UT, USA
| | - Umang Swami
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah Salt Lake, UT, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah Salt Lake, UT, USA
| |
Collapse
|
29
|
Martinez RS, Salji MJ, Rushworth L, Ntala C, Rodriguez Blanco G, Hedley A, Clark W, Peixoto P, Hervouet E, Renaude E, Kung SHY, Galbraith LCA, Nixon C, Lilla S, MacKay GM, Fazli L, Gaughan L, Sumpton D, Gleave ME, Zanivan S, Blomme A, Leung HY. SLFN5 Regulates LAT1-Mediated mTOR Activation in Castration-Resistant Prostate Cancer. Cancer Res 2021; 81:3664-3678. [PMID: 33985973 DOI: 10.1158/0008-5472.can-20-3694] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/15/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022]
Abstract
Androgen deprivation therapy (ADT) is the standard of care for treatment of nonresectable prostate cancer. Despite high treatment efficiency, most patients ultimately develop lethal castration-resistant prostate cancer (CRPC). In this study, we performed a comparative proteomic analysis of three in vivo, androgen receptor (AR)-responsive orthograft models of matched hormone-naïve prostate cancer and CRPC. Differential proteomic analysis revealed that distinct molecular mechanisms, including amino acid (AA) and fatty acid metabolism, are involved in the response to ADT in the different models. Despite this heterogeneity, Schlafen family member 5 (SLFN5) was identified as an AR-regulated protein in CRPC. SLFN5 expression was high in CRPC tumors and correlated with poor patient outcome. In vivo, SLFN5 depletion strongly impaired tumor growth in castrated conditions. Mechanistically, SLFN5 interacted with ATF4 and regulated the expression of LAT1, an essential AA transporter. Consequently, SLFN5 depletion in CRPC cells decreased intracellular levels of essential AA and impaired mTORC1 signaling in a LAT1-dependent manner. These results confirm that these orthograft models recapitulate the high degree of heterogeneity observed in patients with CRPC and further highlight SLFN5 as a clinically relevant target for CRPC. SIGNIFICANCE: This study identifies SLFN5 as a novel regulator of the LAT1 amino acid transporter and an essential contributor to mTORC1 activity in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Rafael S Martinez
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Mark J Salji
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Linda Rushworth
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Chara Ntala
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | | | - Ann Hedley
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - William Clark
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Paul Peixoto
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- EPIGENExp, (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | - Eric Hervouet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- EPIGENExp, (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | - Elodie Renaude
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- EPIGENExp, (EPIgenetics and GENe EXPression Technical Platform), Besançon, France
| | - Sonia H Y Kung
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Laura C A Galbraith
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Colin Nixon
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Sergio Lilla
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Gillian M MacKay
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Ladan Fazli
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Luke Gaughan
- Northern Institute for Cancer Research, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Sumpton
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Martin E Gleave
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Sara Zanivan
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Arnaud Blomme
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom.
| | - Hing Y Leung
- CRUK Beatson Institute, Garscube Estate, Glasgow, United Kingdom.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
30
|
Lobo J, Constâncio V, Guimarães-Teixeira C, Leite-Silva P, Miranda-Gonçalves V, Sequeira JP, Pistoni L, Guimarães R, Cantante M, Braga I, Maurício J, Looijenga LHJ, Henrique R, Jerónimo C. Promoter methylation of DNA homologous recombination genes is predictive of the responsiveness to PARP inhibitor treatment in testicular germ cell tumors. Mol Oncol 2021; 15:846-865. [PMID: 33513287 PMCID: PMC8024740 DOI: 10.1002/1878-0261.12909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Testicular germ cell tumors (TGCTs) are the most common cancers in men aged 15-39 years and are divided into two major groups, seminomas and nonseminomas. Novel treatment options are required for these patients, to limit side effects of chemotherapy. We hypothesized that promoter methylation of relevant homologous recombination (HR) genes might be predictive of response to poly-ADP ribose polymerase inhibitors (PARPis) in TGCTs. We report a study pipeline combining in silico, in vitro, and clinical steps. By using several databases and in silico tools, we identified BRCA1, RAD51C, PALB2, RAD54B, and SYCP3 as the most relevant genes for further investigation and pinpointed specific CpG sites with pronounced negative correlation to gene expression. Nonseminomas displayed significantly higher methylation levels for all target genes, where increased methylation was observed in patients with more differentiated subtypes and higher disease burden. We independently performed second-line targeted validation in tissue series from TGCT patients. A moderate and/or strong anti-correlation between gene expression (assessed by RNA-sequencing) and promoter methylation (assessed by 450k array) was found, for all of the targets. As a proof of concept, we demonstrated the sensitivity of TGCT cell lines to Olaparib, which associated with differential methylation levels of a subset of targets, namely BRCA1 and RAD51C. Our findings support the use of HR genes promoter methylation as a predictor of the therapeutic response to PARPis in patients with TGCT.
Collapse
Affiliation(s)
- João Lobo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Portugal.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Vera Constâncio
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Catarina Guimarães-Teixeira
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Pedro Leite-Silva
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Vera Miranda-Gonçalves
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - José Pedro Sequeira
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal
| | - Laura Pistoni
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Biology, University of Pisa, Italy
| | - Rita Guimarães
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | - Mariana Cantante
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | - Isaac Braga
- Department of Urology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | - Joaquina Maurício
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPOP), Portugal
| | | | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P, CCC), Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Portugal
| |
Collapse
|
31
|
Siva N, Gupta S, Gupta A, Shukla JN, Malik B, Shukla N. Genome-editing approaches and applications: a brief review on CRISPR technology and its role in cancer. 3 Biotech 2021; 11:146. [PMID: 33732568 PMCID: PMC7910401 DOI: 10.1007/s13205-021-02680-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023] Open
Abstract
The development of genome-editing technologies in 1970s has discerned a new beginning in the field of science. Out of different genome-editing approaches such as Zing-finger nucleases, TALENs, and meganucleases, clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 (CRISPR/Cas9) is a recent and versatile technology that has the ability of making changes to the genome of different organisms with high specificity. Cancer is a complex process that is characterized by multiple genetic and epigenetic changes resulting in abnormal cell growth and proliferation. As cancer is one of the leading causes of deaths worldwide, a large number of studies are done to understand the molecular mechanisms underlying the development of cancer. Because of its high efficiency and specificity, CRISPR/Cas9 has emerged as a novel and powerful tool in the field of cancer research. CRISPR/Cas9 has the potential to accelerate cancer research by dissecting tumorigenesis process, generating animal and cellular models, and identify drug targets for chemotherapeutic approaches. However, despite having tremendous potential, there are certain challenges associated with CRISPR/Cas9 such as safe delivery to the target, potential off-target effects and its efficacy which needs to be addressed prior to its clinical application. In this review, we give a gist of different genome-editing technologies with a special focus on CRISPR/Cas9 development, its mechanism of action and its applications, especially in different type of cancers. We also highlight the importance of CRISPR/Cas9 in generating animal models of different cancers. Finally, we present an overview of the clinical trials and discuss the challenges associated with translating CRISPR/Cas9 in clinical use.
Collapse
Affiliation(s)
- Narmadhaa Siva
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Statue Circle, Jaipur, India
| | - Sonal Gupta
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Statue Circle, Jaipur, India
| | - Ayam Gupta
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Statue Circle, Jaipur, India
| | - Jayendra Nath Shukla
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Bandarsindari, Ajmer, India
| | - Babita Malik
- Department of Chemistry, Manipal University Jaipur, Jaipur, India
| | - Nidhi Shukla
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Statue Circle, Jaipur, India
- Department of Chemistry, Manipal University Jaipur, Jaipur, India
| |
Collapse
|
32
|
Katleba K, Lombard AP, Tsamouri MM, Baek HB, Nishida KS, Libertini SJ, Platero AJ, Ma AH, Pan CX, Ghosh PM, Mudryj M. Depletion of androgen receptor low molecular weight isoform reduces bladder tumor cell viability and induces apoptosis. Cancer Lett 2021; 504:49-57. [PMID: 33549708 DOI: 10.1016/j.canlet.2021.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 01/21/2023]
Abstract
Bladder cancer (BlCa) exhibits a gender disparity where men are three times more likely to develop the malignancy than women suggesting a role for the androgen receptor (AR). Here we report that BlCa cells express low molecular weight (LMW) AR isoforms that are missing the ligand binding domain (LBD). Isoform expression was detected in most BlCa cells, while a few express the full-length AR. Immunofluorescence studies detect AR in the nucleus and cytoplasm, and localization is cell dependent. Cells with nuclear AR expression exhibit reduced viability and increased apoptosis on total AR depletion. A novel AR-LMW variant, AR-v19, that is missing the LBD and contains 15 additional amino acids encoded by intron 3 sequences was detected in most BlCa malignancies. AR-v19 localizes to the nucleus and can transactivate AR-dependent transcription in a dose dependent manner. AR-v19 depletion impairs cell viability and promotes apoptosis in cells that express this variant. Thus, AR splice variant expression is common in BlCa and instrumental in ensuring cell survival. This suggests that targeting AR or AR downstream effectors may be a therapeutic strategy for the treatment of this malignancy.
Collapse
Affiliation(s)
- Kimberley Katleba
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA; Department of Medical Microbiology and Immunology, USA
| | - Alan P Lombard
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA; Department of Medical Microbiology and Immunology, USA; Biochemistry, Molecular, Cellular, and Developmental Biology Graduate Group and Biotechnology Program, USA
| | - Maria-Malvina Tsamouri
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA; Department of Urologic Surgery, University of California, 1 Shields Avenue, UC Davis, Davis, CA, 95616, USA
| | - Han Bit Baek
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA; Department of Medical Microbiology and Immunology, USA
| | | | - Stephen J Libertini
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA; Department of Medical Microbiology and Immunology, USA
| | | | - Ai-Hong Ma
- Department of Urologic Surgery, University of California, 1 Shields Avenue, UC Davis, Davis, CA, 95616, USA
| | - Chong-Xian Pan
- Department of Faculty of Medicine, Harvard Medical School, West Roxbury, MA, 02115, USA
| | - Paramita M Ghosh
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA; Department of Urologic Surgery, University of California, 1 Shields Avenue, UC Davis, Davis, CA, 95616, USA
| | - Maria Mudryj
- Veterans Affairs-Northern California Health Care System, Mather, CA, USA; Department of Medical Microbiology and Immunology, USA.
| |
Collapse
|
33
|
A detailed characterization of stepwise activation of the androgen receptor variant 7 in prostate cancer cells. Oncogene 2020; 40:1106-1117. [PMID: 33323969 PMCID: PMC7880901 DOI: 10.1038/s41388-020-01585-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/11/2020] [Accepted: 11/24/2020] [Indexed: 01/28/2023]
Abstract
Expression of the andrgogen receptor splice variant 7 (AR-V7) is frequently detected in castrate resistant prostate cancer and associated with resistance to AR-targeted therapies. While we have previously noted that homodimerization is required for the transcriptional activity of AR-V7 and that AR-V7 can also form heterodimers with the full-length AR (AR-FL), there are still many gaps of knowledge in AR-V7 stepwise activation. In the present study, we show that neither AR-V7 homodimerization nor AR-V7/AR-FL heterodimerization requires cofactors or DNA binding. AR-V7 can enter the nucleus as a monomer and drive a transcriptional program and DNA-damage repair as a homodimer. While forming a heterodimer with AR-FL to induce nuclear localization of unliganded AR-FL, AR-V7 does not need to interact with AR-FL to drive gene transcription or DNA-damage repair in prostate cancer cells that co-express AR-V7 and AR-FL. These data indicate that AR-V7 can function independently of its interaction with AR-FL in the true castrate state or “absence of ligand”, providing support for the utility of targeting AR-V7 in improving outcomes of patients with castrate resistant prostate cancer.
Collapse
|
34
|
Wang Y, Chen J, Wu Z, Ding W, Gao S, Gao Y, Xu C. Mechanisms of enzalutamide resistance in castration-resistant prostate cancer and therapeutic strategies to overcome it. Br J Pharmacol 2020; 178:239-261. [PMID: 33150960 DOI: 10.1111/bph.15300] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer is the second most common malignancy in men and androgen deprivation therapy is the first-line therapy. However, most cases will eventually develop castration-resistant prostate cancer after androgen deprivation therapy treatment. Enzalutamide is a second-generation androgen receptor antagonist approved by the Food and Drug Administration to treat patients with castration-resistant prostate cancer. Unfortunately, patients receiving enzalutamide treatment will ultimately develop resistance via various complicated mechanisms. This review examines the emerging information on these resistance mechanisms, including androgen receptor-related signalling pathways, glucocorticoid receptor-related pathways and metabolic effects. Notably, lineage plasticity and phenotype switching, gene polymorphisms and the relationship between microRNAs and drug resistance are addressed. Furthermore, potential therapeutic strategies for enzalutamide-resistant castration-resistant prostate cancer treatment are suggested, which can help discover more effective and specific regimens to overcome enzalutamide resistance.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiyuan Chen
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Zhengjie Wu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shen Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Gao
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
35
|
Powers E, Karachaliou GS, Kao C, Harrison MR, Hoimes CJ, George DJ, Armstrong AJ, Zhang T. Novel therapies are changing treatment paradigms in metastatic prostate cancer. J Hematol Oncol 2020; 13:144. [PMID: 33115529 PMCID: PMC7594418 DOI: 10.1186/s13045-020-00978-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/11/2020] [Indexed: 12/11/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains a terminal diagnosis with an aggressive disease course despite currently approved therapeutics. The recent successful development of poly ADP-ribose polymerase (PARP) inhibitors for patients with mCRPC and mutations in DNA damage repair genes has added to the treatment armamentarium and improved personalized treatments for prostate cancer. Other promising therapeutic agents currently in clinical development include the radiotherapeutic 177-lutetium-prostate-specific membrane antigen (PSMA)-617 targeting PSMA-expressing prostate cancer and combinations of immunotherapy with currently effective treatment options for prostate cancer. Herein, we have highlighted the progress in systemic treatments for mCRPC and the promising agents currently in ongoing clinical trials.
Collapse
Affiliation(s)
- Eric Powers
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Georgia Sofia Karachaliou
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Chester Kao
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Michael R Harrison
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Christopher J Hoimes
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA.,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA. .,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA.
| |
Collapse
|
36
|
Germain L, Lafront C, Beaudette J, Karthik Poluri RT, Weidmann C, Audet-Walsh É. Alternative splicing regulation by the androgen receptor in prostate cancer cells. J Steroid Biochem Mol Biol 2020; 202:105710. [PMID: 32534106 DOI: 10.1016/j.jsbmb.2020.105710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 03/11/2020] [Accepted: 06/02/2020] [Indexed: 11/19/2022]
Abstract
The androgen receptor (AR) is a transcription factor that drives prostate cancer (PCa) by modulating the expression of thousands of genes to promote proliferation and survival and to reprogram metabolism. However, how AR activation controls alternative splicing is mostly unknown. Our objective was to define its role in the transcriptome-wide regulation of alternative splicing. Three human PCa models-LNCaP, LAPC4, and 22Rv1 cells-were treated with and without androgens, and RNA was purified for deep-sequencing analyses (RNA-seq). Several bioinformatic tools were then used to study alternative splicing. We demonstrate that in the absence of androgens, alternative splicing complexity is similar among AR-positive PCa cells, with 48 % of all transcripts having various levels of alternative splicing. We also describe alternative splicing differences among cell lines, such as specific splicing of AR, REST, TSC2, and CTBP1. Interestingly, AR activation changed the alternative splicing of thousands of genes in all the PCa cell lines tested. Overlap between AR-sensitive alternative splicing events revealed that genes linked to cell metabolism are major targets for this specific modulation. These genes encode metabolic enzymes such as the prostate-specific membrane antigen, encoded by FOLH1, and the malate dehydrogenase 1 (MDH1). Overall, our study presents a comprehensive analysis of the PCa cell transcriptome and its modulation by AR, revealing a significant enrichment of metabolic genes in this AR-dependent regulation of alternative splicing.
Collapse
Affiliation(s)
- Lucas Germain
- Département de biochimie, microbiologie et bio-informatique, Faculté des sciences et de génie, Université Laval, Canada; Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Canada; Centre de recherche sur le cancer de l'Université Laval, Canada
| | - Camille Lafront
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Canada; Centre de recherche sur le cancer de l'Université Laval, Canada; Department of molecular medicine, Faculty of Medicine, Université Laval, Québec City, G1V 0A6, Canada
| | - Jolyane Beaudette
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Canada; Centre de recherche sur le cancer de l'Université Laval, Canada; Department of molecular medicine, Faculty of Medicine, Université Laval, Québec City, G1V 0A6, Canada
| | - Raghavendra Tejo Karthik Poluri
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Canada; Centre de recherche sur le cancer de l'Université Laval, Canada; Department of molecular medicine, Faculty of Medicine, Université Laval, Québec City, G1V 0A6, Canada
| | - Cindy Weidmann
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Canada; Centre de recherche sur le cancer de l'Université Laval, Canada
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Canada; Centre de recherche sur le cancer de l'Université Laval, Canada; Department of molecular medicine, Faculty of Medicine, Université Laval, Québec City, G1V 0A6, Canada.
| |
Collapse
|
37
|
Matos B, Howl J, Jerónimo C, Fardilha M. The disruption of protein-protein interactions as a therapeutic strategy for prostate cancer. Pharmacol Res 2020; 161:105145. [PMID: 32814172 DOI: 10.1016/j.phrs.2020.105145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is one of the most common male-specific cancers worldwide, with high morbidity and mortality rates associated with advanced disease stages. The current treatment options of PCa are prostatectomy, hormonal therapy, chemotherapy or radiotherapy, the selection of which is usually dependent upon the stage of the disease. The development of PCa to a castration-resistant phenotype (CRPC) is associated with a more severe prognosis requiring the development of a new and effective therapy. Protein-protein interactions (PPIs) have been recognised as an emerging drug modality and targeting PPIs is a promising therapeutic approach for several diseases, including cancer. The efficacy of several compounds in which target PPIs and consequently impair disease progression were validated in phase I/II clinical trials for different types of cancer. In PCa, various small molecules and peptides proved successful in inhibiting important PPIs, mainly associated with the androgen receptor (AR), Bcl-2 family proteins, and kinases/phosphatases, thus impairing the growth of PCa cells in vitro. Moreover, a majority of these compounds require further validation in vivo and, preferably, in clinical trials. In addition, several other PPIs associated with PCa progression have been identified and now require experimental validation as potential therapeutic loci. In conclusion, we consider the disruption of PPIs to be a promising though challenging therapeutic strategy for PCa. Agents which modulate PPIs might be employed as a monotherapy or as an adjunct to classical chemotherapeutics to overcome drug resistance and improve efficacy. The discovery of new PPIs with important roles in disease progression, and of novel optimized strategies to target them are major challenges for the scientific and pharmacological communities.
Collapse
Affiliation(s)
- Bárbara Matos
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal
| | - John Howl
- Molecular Pharmacology Group, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), Research Center-LAB 3, F Bdg., 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar- University of Porto (ICBAS-UP), Porto, Portugal
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
38
|
Hirayama Y, Tam T, Jian K, Andersen RJ, Sadar MD. Combination therapy with androgen receptor N-terminal domain antagonist EPI-7170 and enzalutamide yields synergistic activity in AR-V7-positive prostate cancer. Mol Oncol 2020; 14:2455-2470. [PMID: 32734688 PMCID: PMC7530779 DOI: 10.1002/1878-0261.12770] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/08/2020] [Accepted: 07/23/2020] [Indexed: 11/09/2022] Open
Abstract
Resistance of castration-resistant prostate cancer (CRPC) to enzalutamide and abiraterone involves the expression of constitutively active, truncated androgen receptor (AR) splice variants (AR-Vs) that lack a C-terminal ligand-binding domain (LBD). Both full-length AR and truncated AR-Vs require a functional N-terminal domain (NTD) for transcriptional activity thereby providing rationale for the development of ralaniten (EPI-002) as a first-in-class antagonist of the AR-NTD. Here, we evaluated the antitumor effect of a next-generation analog of ralaniten (EPI-7170) as a monotherapy or in combination with enzalutamide in prostate cancer cells that express AR-V7 that were resistant to enzalutamide. EPI-7170 had 8-9 times improved potency compared to ralaniten. Enzalutamide increased levels of AR-V7 and expression of its target genes. Knockdown of AR-V7 restored sensitivity to enzalutamide, indicating a role for AR-V7 in the mechanism of resistance. EPI-7170 inhibited expression of genes transcriptionally regulated by full-length AR and AR-V7. A combination of EPI-7170 and enzalutamide resulted in synergistic inhibition of proliferation of enzalutamide-resistant cells that was consistent with results from cell cycle and clonogenic assays. In addition, this drug enhanced the antitumor effect of enzalutamide in enzalutamide-resistant CRPC preclinical models. Thus, a combination therapy targeting both the NTD and LBD of AR, and thereby blocking both full-length AR and AR-Vs, has potential for the treatment of enzalutamide-resistant CRPC.
Collapse
Affiliation(s)
| | - Teresa Tam
- Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Kunzhong Jian
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada.,Department of Earth, Ocean, and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Raymond J Andersen
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada.,Department of Earth, Ocean, and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Marianne D Sadar
- Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
39
|
Hankey W, Chen Z, Wang Q. Shaping Chromatin States in Prostate Cancer by Pioneer Transcription Factors. Cancer Res 2020; 80:2427-2436. [PMID: 32094298 PMCID: PMC7299826 DOI: 10.1158/0008-5472.can-19-3447] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/14/2020] [Accepted: 02/19/2020] [Indexed: 01/28/2023]
Abstract
The androgen receptor (AR) is a critical therapeutic target in prostate cancer that responds to antagonists in primary disease, but inevitably becomes reactivated, signaling onset of the lethal castration-resistant prostate cancer (CRPC) stage. Epigenomic investigation of the chromatin environment and interacting partners required for AR transcriptional activity has uncovered three pioneer factors that open up chromatin and facilitate AR-driven transcriptional programs. FOXA1, HOXB13, and GATA2 are required for normal AR transcription in prostate epithelial development and for oncogenic AR transcription during prostate carcinogenesis. AR signaling is dependent upon these three pioneer factors both before and after the clinical transition from treatable androgen-dependent disease to untreatable CRPC. Agents targeting their respective DNA binding or downstream chromatin-remodeling events have shown promise in preclinical studies of CRPC. AR-independent functions of FOXA1, HOXB13, and GATA2 are emerging as well. While all three pioneer factors exert effects that promote carcinogenesis, some of their functions may inhibit certain stages of prostate cancer progression. In all, these pioneer factors represent some of the most promising potential therapeutic targets to emerge thus far from the study of the prostate cancer epigenome.
Collapse
Affiliation(s)
- William Hankey
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| | - Zhong Chen
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina.
| | - Qianben Wang
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
40
|
Selective targeting of PARP-2 inhibits androgen receptor signaling and prostate cancer growth through disruption of FOXA1 function. Proc Natl Acad Sci U S A 2019; 116:14573-14582. [PMID: 31266892 DOI: 10.1073/pnas.1908547116] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Androgen receptor (AR) is a ligand-activated transcription factor and a key driver of prostate cancer (PCa) growth and progression. Understanding the factors influencing AR-mediated gene expression provides new opportunities for therapeutic intervention. Poly(ADP-ribose) Polymerase (PARP) is a family of enzymes, which posttranslationally modify a range of proteins and regulate many different cellular processes. PARP-1 and PARP-2 are two well-characterized PARP members, whose catalytic activity is induced by DNA-strand breaks and responsible for multiple DNA damage repair pathways. PARP inhibitors are promising therapeutic agents that show synthetic lethality against many types of cancer (including PCa) with homologous recombination (HR) DNA-repair deficiency. Here, we show that, beyond DNA damage repair function, PARP-2, but not PARP-1, is a critical component in AR transcriptional machinery through interacting with the pioneer factor FOXA1 and facilitating AR recruitment to genome-wide prostate-specific enhancer regions. Analyses of PARP-2 expression at both mRNA and protein levels show significantly higher expression of PARP-2 in primary PCa tumors than in benign prostate tissues, and even more so in castration-resistant prostate cancer (CRPC) tumors. Selective targeting of PARP-2 by genetic or pharmacological means blocks interaction between PARP-2 and FOXA1, which in turn attenuates AR-mediated gene expression and inhibits AR-positive PCa growth. Next-generation antiandrogens act through inhibiting androgen synthesis (abiraterone) or blocking ligand binding (enzalutamide). Selective targeting of PARP-2, however, may provide an alternative therapeutic approach for AR inhibition by disruption of FOXA1 function, which may be beneficial to patients, irrespective of their DNA-repair deficiency status.
Collapse
|
41
|
Mota STS, Vecchi L, Zóia MAP, Oliveira FM, Alves DA, Dornelas BC, Bezerra SM, Andrade VP, Maia YCP, Neves AF, Goulart LR, Araújo TG. New Insights into the Role of Polybromo-1 in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20122852. [PMID: 31212728 PMCID: PMC6627401 DOI: 10.3390/ijms20122852] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 01/22/2023] Open
Abstract
The human protein Polybromo-1 (PBMR1/BAF180) is a component of the SWI/SNF chromatin-remodeling complex that has been reported to be deregulated in tumors. However, its role in prostate cancer (PCa) is largely unknown. In this study, we described the PBRM1 transcriptional levels and the protein expression/localization in tissues of PCa patients and in prostatic cell lines. Increased PBRM1 mRNA levels were found in PCa samples, when compared to benign disease, and were correlated with higher Gleason score. We also verified that only the nuclear localization of PBRM1 protein is correlated with a more aggressive disease and high Prostate-Specific Antigen (PSA) levels in tissue microarrays. Intriguing expression patterns of mRNA and protein were identified in the cell lines. Although PBRM1 protein was restricted to the nuclei, in tumor cell lines in non-neoplastic cells, it was also present in vesicular-like structures that were dispersed within the cytoplasm. We knocked-down PBRM1 in the castration-resistant PCa (CRPC) cell line PC-3 and we verified that PBRM1 promotes the expression of several markers of aggressiveness, including EpCAM, TGF-β, and N-Cadherin. Therefore, our data supported the hypothesis that PBRM1 displays a pivotal role in the promotion and maintenance of the malignant behavior of PCa, especially in CRPC.
Collapse
Affiliation(s)
- Sara T S Mota
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Lara Vecchi
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Mariana A P Zóia
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Fabrícia M Oliveira
- Faculty of Mathematics, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
| | - Douglas A Alves
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Bruno C Dornelas
- Pathology Division, Internal Medicine, University Hospital, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | | | | | - Yara C P Maia
- Medical Faculty, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Adriana F Neves
- Laboratory of Molecular Biology, Federal University of Goias-GO, Goiânia-GO 75704-020, Brazil.
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
- University of California Davis, Department of Medical Microbiology and Immunology, Davis, CA 95616, USA.
| | - Thaise G Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| |
Collapse
|