1
|
Yuan H, Cai R, Chen B, Wang Q, Wang M, An J, An W, Tao Y, Yu J, Jiang B, Zhang Y, Xu M. Acetylated KHSRP impairs DNA-damage-response-related mRNA decay and facilitates prostate cancer tumorigenesis. Mol Oncol 2024; 18:2314-2330. [PMID: 38501452 PMCID: PMC11467790 DOI: 10.1002/1878-0261.13634] [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: 10/22/2023] [Revised: 01/28/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Androgen-regulated DNA damage response (DDR) is one of the essential mechanisms in prostate cancer (PCa), a hormone-sensitive disease. The heterogeneous nuclear ribonucleoprotein K (hnRNPK)-homology splicing regulatory protein known as far upstream element-binding protein 2 (KHSRP) is an RNA-binding protein that can attach to AU-rich elements in the 3' untranslated region (3'-UTR) of messenger RNAs (mRNAs) to mediate mRNA decay and emerges as a critical regulator in the DDR to preserve genome integrity. Nevertheless, how KHSRP responds to androgen-regulated DDR in PCa development remains unclear. This study found that androgen can significantly induce acetylation of KHSRP, which intrinsically drives tumor growth in xenografted mice. Moreover, enhanced KHSRP acetylation upon androgen stimuli impedes KHSRP-regulated DDR gene expression, as seen by analyzing RNA sequencing (RNA-seq) and Gene Set Enrichment Analysis (GSEA) datasets. Additionally, NAD-dependent protein deacetylase sirtuin-7 (SIRT7) is a promising deacetylase of KHSRP, and androgen stimuli impairs its interaction with KHSRP to sustain the increased KHSRP acetylation level in PCa. We first report the acetylation of KHSRP induced by androgen, which interrupts the KHSRP-regulated mRNA decay of the DDR-related genes to promote the tumorigenesis of PCa. This study provides insight into KHSRP biology and potential therapeutic strategies for PCa treatment, particularly that of castration-resistant PCa.
Collapse
Affiliation(s)
- Haihua Yuan
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Renjie Cai
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Biying Chen
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Qian Wang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Mengting Wang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Junyi An
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Weishu An
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Ye Tao
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Bin Jiang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Yanjie Zhang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Ming Xu
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| |
Collapse
|
2
|
Akhoundova D, Francica P, Rottenberg S, Rubin MA. DNA Damage Response and Mismatch Repair Gene Defects in Advanced and Metastatic Prostate Cancer. Adv Anat Pathol 2024; 31:61-69. [PMID: 38008971 PMCID: PMC10846598 DOI: 10.1097/pap.0000000000000422] [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] [Indexed: 11/28/2023]
Abstract
Alterations in DNA damage response (DDR) and related genes are present in up to 25% of advanced prostate cancers (PCa). Most frequently altered genes are involved in the homologous recombination repair, the Fanconi anemia, and the mismatch repair pathways, and their deficiencies lead to a highly heterogeneous spectrum of DDR-deficient phenotypes. More than half of these alterations concern non- BRCA DDR genes. From a therapeutic perspective, poly-ADP-ribose polymerase inhibitors have demonstrated robust clinical efficacy in tumors with BRCA2 and BRCA1 alterations. Mismatch repair-deficient PCa, and a subset of CDK12-deficient PCa, are vulnerable to immune checkpoint inhibitors. Emerging data point to the efficacy of ATR inhibitors in PCa with ATM deficiencies. Still, therapeutic implications are insufficiently clarified for most of the non- BRCA DDR alterations, and no successful targeted treatment options have been established.
Collapse
Affiliation(s)
- Dilara Akhoundova
- Department for BioMedical Research
- Department of Medical Oncology
- Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Paola Francica
- Department for BioMedical Research
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern
- Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Sven Rottenberg
- Department for BioMedical Research
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern
- Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Mark A. Rubin
- Department for BioMedical Research
- Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Hasterok S, Scott TG, Roller DG, Spencer A, Dutta AB, Sathyan KM, Frigo DE, Guertin MJ, Gioeli D. The Androgen Receptor Does Not Directly Regulate the Transcription of DNA Damage Response Genes. Mol Cancer Res 2023; 21:1329-1341. [PMID: 37698543 PMCID: PMC11022999 DOI: 10.1158/1541-7786.mcr-23-0358] [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: 05/14/2023] [Revised: 08/02/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
The clinical success of combined androgen deprivation therapy (ADT) and radiotherapy (RT) in prostate cancer created interest in understanding the mechanistic links between androgen receptor (AR) signaling and the DNA damage response (DDR). Convergent data have led to a model where AR both regulates, and is regulated by, the DDR. Integral to this model is that the AR regulates the transcription of DDR genes both at a steady state and in response to ionizing radiation (IR). In this study, we sought to determine which immediate transcriptional changes are induced by IR in an AR-dependent manner. Using PRO-seq to quantify changes in nascent RNA transcription in response to IR, the AR antagonist enzalutamide, or the combination of the two, we find that enzalutamide treatment significantly decreased expression of canonical AR target genes but had no effect on DDR gene sets in prostate cancer cells. Surprisingly, we also found that the AR is not a primary regulator of DDR genes either in response to IR or at a steady state in asynchronously growing prostate cancer cells. IMPLICATIONS Our data indicate that the clinical benefit of combining ADT with RT is not due to direct AR regulation of DDR gene transcription, and that the field needs to consider alternative mechanisms for this clinical benefit.
Collapse
Affiliation(s)
- Sylwia Hasterok
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Thomas G. Scott
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
| | - Devin G. Roller
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Adam Spencer
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Arun B. Dutta
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
| | - Kizhakke M Sathyan
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut, Farmington, Connecticut 06030, USA
| | - Daniel E. Frigo
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Michael J. Guertin
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut, Farmington, Connecticut 06030, USA
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, Connecticut 06030, USA
| | - Daniel Gioeli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Cancer Center Member, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
4
|
Giesen A, Baekelandt L, Devlies W, Devos G, Dumez H, Everaerts W, Claessens F, Joniau S. Double trouble for prostate cancer: synergistic action of AR blockade and PARPi in non-HRR mutated patients. Front Oncol 2023; 13:1265812. [PMID: 37810962 PMCID: PMC10551452 DOI: 10.3389/fonc.2023.1265812] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men worldwide. Despite better and more intensive treatment options in earlier disease stages, a large subset of patients still progress to metastatic castration-resistant PCa (mCRPC). Recently, poly-(ADP-ribose)-polymerase (PARP)-inhibitors have been introduced in this setting. The TALAPRO-2 and PROpel trials both showed a marked benefit of PARPi in combination with an androgen receptor signaling inhibitor (ARSI), compared with an ARSI alone in both the homologous recombination repair (HRR)-mutated, as well as in the HRR-non-mutated subgroup. In this review, we present a comprehensive overview of how maximal AR-blockade via an ARSI in combination with a PARPi has a synergistic effect at the molecular level, leading to synthetic lethality in both HRR-mutated and HRR-non-mutated PCa patients. PARP2 is known to be a cofactor of the AR complex, needed for decompacting the chromatin and start of transcription of AR target genes (including HRR genes). The inhibition of PARP thus reinforces the effect of an ARSI. The deep androgen deprivation caused by combining androgen deprivation therapy (ADT) with an ARSI, induces an HRR-like deficient state, often referred to as "BRCA-ness". Further, PARPi will prevent the repair of single-strand DNA breaks, leading to the accumulation of DNA double-strand breaks (DSBs). Due to the induced HRR-deficient state, DSBs cannot be repaired, leading to apoptosis.
Collapse
Affiliation(s)
- Alexander Giesen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Loïc Baekelandt
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, Catholic University Leuven (KU Leuven), Leuven, Belgium
| | - Gaëtan Devos
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Herlinde Dumez
- Department of Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Frank Claessens
- Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, Catholic University Leuven (KU Leuven), Leuven, Belgium
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, Catholic University Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
5
|
Chen D, Chou FJ, Chen Y, Huang CP, Tian H, Wang Y, Niu Y, You B, Yeh S, Xing N, Chang C. Targeting the radiation-induced ARv7-mediated circNHS/miR-512-5p/XRCC5 signaling with Quercetin increases prostate cancer radiosensitivity. J Exp Clin Cancer Res 2022; 41:235. [PMID: 35918767 PMCID: PMC9347162 DOI: 10.1186/s13046-022-02287-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 02/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background Radiation therapy (RT) with androgen deprivation therapy (ADT) is an effective therapy to suppress the locally advanced prostate cancer (PCa). However, we unexpectedly found that RT could also induce the androgen receptor splice variant 7 (ARv7) expression to decrease the radiosensitivity. Methods The study was designed to target ARv7 expression with Quercetin or ARv7-shRNA that leads to enhancing and increasing the radiation sensitivity to better suppress the PCa that involved the modulation of the circNHS/miR-512-5p/XRCC5 signaling. Results Mechanism studies revealed that RT-induced ARv7 may function via altering the circNHS/miR-512-5p/XRCC5 signaling to decrease the radiosensitivity. Results from preclinical studies using multiple in vitro cell lines and in vivo mouse models concluded that combining RT with the small molecule of Quercetin to target full-length AR and ARv7 could lead to better efficacy to suppress PCa progression. Conclusion Together, these results suggest that ARv7 may play key roles to alter the PCa radiosensitivity, and targeting this newly identified ARv7 mediated circNHS/miR-512-5p/XRCC5 signaling with Quercetin may help physicians to develop a novel RT to better suppress the progression of PCa. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02287-4.
Collapse
|
6
|
Iwanaga M, Kawamura H, Kubo N, Mizukami T, Oike T, Sato H, Miyazawa Y, Sekine Y, Kawabata-Iwakawa R, Nishiyama M, Ohno T, Nakano T. Double-layer omics analysis of castration- and X-ray-resistant prostate cancer cells. JOURNAL OF RADIATION RESEARCH 2022; 63:585-590. [PMID: 35589101 PMCID: PMC9303586 DOI: 10.1093/jrr/rrac022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 01/31/2022] [Indexed: 06/15/2023]
Abstract
Castration-resistant prostate cancer shows resistance to not only androgen deprivation therapy (ADT) but also X-ray therapy. On the other hand, carbon ion beams have a high biological effect and are used for various cancers showing resistance to X-ray therapy. The purposes of this study are to clarify the difference in the sensitivity of Castration-resistant prostate cancer to X-ray and carbon ion beams and to elucidate the mechanism. The androgen-insensitive prostate cancer cell line LNCaP-LA established by culturing the androgen-sensitive prostate cancer cell line LNCaP for 2 years in androgen-free medium was used for this study. First, colony formation assays were performed to investigate its sensitivity to X-ray and carbon ion beams. Next, DNA mutation analysis on 409 cancer-related genes and comprehensive transcriptome analysis (RNA-seq) were performed with a next-generation sequencer. Lethal dose 50 values of X-rays for LNCaP and LNCaP-LA were 1.4 Gy and 2.8 Gy, respectively (P < 0.01). The Lethal dose 50 values of carbon ion beams were 0.9 Gy and 0.7 Gy, respectively (P = 0.09). On DNA mutation analysis, AR mutation was observed specifically in LNCaP-LA. From RNA-seq, 181 genes were identified as differentially expressed genes (DEGs; FDR <0.10, P < 0.00076) between LNCaP and LNCaP-LA. Function analysis suggested that cell death was suppressed in LNCaP-LA, and pathway analysis suggested that the NRF2-pathway involved in intracellular oxidative stress prevention was activated in LNCaP-LA. LNCaP-LA showed X-ray resistance compared to LNCaP and sensitivity to carbon ion beams. The AR mutation and the NRF2-pathway were suggested as causes of resistance.
Collapse
Affiliation(s)
- Mototaro Iwanaga
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hidemasa Kawamura
- Corresponding author. 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, JAPAN, Phone: +81-27-220-8383, Fax: +81-27-220-8397, E-mail:
| | - Nobuteru Kubo
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tatsuji Mizukami
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takahiro Oike
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiro Sato
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yoshiyuki Miyazawa
- Department of Urology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yoshitaka Sekine
- Department of Urology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Masahiko Nishiyama
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
- Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tatsuya Ohno
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
7
|
Saxby H, Boussios S, Mikropoulos C. Androgen Receptor Gene Pathway Upregulation and Radiation Resistance in Oligometastatic Prostate Cancer. Int J Mol Sci 2022; 23:ijms23094786. [PMID: 35563176 PMCID: PMC9105839 DOI: 10.3390/ijms23094786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/09/2022] [Accepted: 04/24/2022] [Indexed: 12/20/2022] Open
Abstract
Stereotactic ablative body radiotherapy (SABR) is currently used as a salvage intervention for men with oligometastatic prostate cancer (PC), and increasingly so since the results of the Stereotactic Ablative Body Radiotherapy for the Comprehensive Treatment of Oligometastatic Cancers (SABR-COMET) trial reported a significant improvement in overall survival with SABR. The addition of androgen deprivation therapy (ADT) to localised prostate radiotherapy improves survival as it sensitises PC to radiotherapy-induced cell death. The importance of the androgen receptor (AR) gene pathway in the development of resistance to radiotherapy is well established. In this review paper, we will examine the data to determine how we can overcome the upregulation of the AR pathway and suggest a strategy for improving outcomes in men with oligometastatic hormone-sensitive PC.
Collapse
Affiliation(s)
- Helen Saxby
- Torbay & South Devon NHS Healthcare Foundation Trust, Lowes Bridge, Torquay TQ2 7AA, UK;
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham Kent ME7 5NY, UK
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki–Thermi, 57001 Thessaloniki, Greece
- Correspondence: , or
| | - Christos Mikropoulos
- St Lukes Cancer Centre, Royal Surrey County Hospital, Egerton Rd, Guildford GU2 7XX, UK;
| |
Collapse
|
8
|
Neoadjuvant hormonal therapy before radical prostatectomy in high-risk prostate cancer. Nat Rev Urol 2021; 18:739-762. [PMID: 34526701 DOI: 10.1038/s41585-021-00514-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 02/08/2023]
Abstract
Patients with high-risk prostate cancer treated with curative intent are at an increased risk of biochemical recurrence, metastatic progression and cancer-related death compared with patients treated for low-risk or intermediate-risk disease. Thus, these patients often need multimodal therapy to achieve complete disease control. Over the past two decades, multiple studies on the use of neoadjuvant treatment have been performed using conventional androgen deprivation therapy, which comprises luteinizing hormone-releasing hormone agonists or antagonists and/or first-line anti-androgens. However, despite results from these studies demonstrating a reduction in positive surgical margins and tumour volume, no benefit has been observed in hard oncological end points, such as cancer-related death. The introduction of potent androgen receptor signalling inhibitors (ARSIs), such as abiraterone, apalutamide, enzalutamide and darolutamide, has led to a renewed interest in using neoadjuvant hormonal treatment in high-risk prostate cancer. The addition of ARSIs to androgen deprivation therapy has demonstrated substantial survival benefits in the metastatic castration-resistant, non-metastatic castration-resistant and metastatic hormone-sensitive settings. Intuitively, a similar survival effect can be expected when applying ARSIs as a neoadjuvant strategy in high-risk prostate cancer. Most studies on neoadjuvant ARSIs use a pathological end point as a surrogate for long-term oncological outcome. However, no consensus yet exists regarding the ideal definition of pathological response following neoadjuvant hormonal therapy and pathologists might encounter difficulties in determining pathological response in hormonally treated prostate specimens. The neoadjuvant setting also provides opportunities to gain insight into resistance mechanisms against neoadjuvant hormonal therapy and, consequently, to guide personalized therapy.
Collapse
|
9
|
Qian D, Li Q, Zhu Y, Li D. Comprehensive Analysis of Key Proteins Involved in Radioresistance of Prostate Cancer by Integrating Protein-protein Interaction Networks. Curr Bioinform 2021. [DOI: 10.2174/1574893615999200605143510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Radioresistance remains a significant obstacle in the treatment of prostate
cancer (PCa). The mechanisms underlying the radioresistance in PCa remained to be further
investigated.
Methods:
GSE53902 dataset was used in this study to identify radioresistance-related mRNAs.
Protein-protein interaction (PPI) network was constructed based on STRING analysis. DAVID
system was used to predict the potential roles of radioresistance-related mRNAs.
Results:
We screened and re-annotated the GSE53902 dataset to identify radioresistance-related
mRNAs. A total of 445 up-regulated and 1036 down-regulated mRNAs were identified in
radioresistance PCa cells. Three key PPI networks consisting of 81 proteins were further constructed
in PCa. Bioinformatics analysis revealed that these genes were involved in regulating MAP kinase
activity, response to hypoxia, regulation of the apoptotic process, mitotic nuclear division, and
regulation of mRNA stability. Moreover, we observed that radioresistance-related mRNAs, such as
PRC1, RAD54L, PIK3R3, ASB2, FBXO32, LPAR1, RNF14, and UBA7, were dysregulated and
correlated to the survival time in PCa.
Conclusion:
We thought this study would be useful to understand the mechanisms underlying
radioresistance of PCa and identify novel prognostic markers for PCa.
Collapse
Affiliation(s)
- Duocheng Qian
- Department of Urology, Shanghai Fourth People’s Hospital, Shanghai, 200081, China
| | - Quan Li
- Department of Urology, Shanghai Fourth People’s Hospital, Shanghai, 200081, China
| | - Yansong Zhu
- Department of Urology, Shanghai Fourth People’s Hospital, Shanghai, 200081, China
| | - Dujian Li
- Department of Urology, Shanghai Fourth People’s Hospital, Shanghai, 200081, China
| |
Collapse
|
10
|
The role of regulatory T cells in the pathogenesis and treatment of prostate cancer. Life Sci 2021; 284:119132. [PMID: 33513396 DOI: 10.1016/j.lfs.2021.119132] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Despite developments in the treatment of various cancers, prostate cancer is one of the deadliest diseases known to men. Systemic therapies such as androgen deprivation, chemotherapy, and radiation therapy have not been very successful in treating this disease. Numerous studies have shown that there is a direct relationship between cancer progression and inhibition of anti-tumor immune responses that can lead to progression of various malignancies, including prostate cancer. Interestingly, CD4+CD25+FoxP3+ regulatory T cells significantly accumulate and increase in draining lymph nodes and PBMCs of patients with prostate cancer and other solid tumors. In vivo and in vitro studies have shown that Tregs can suppress anti-tumor responses, which is directly related to the increased risk of cancer recurrence. Tregs are essential for preserving self-tolerance and inhibiting extra immune responses harmful to the host. Since the tumor-related antigens are mainly self-antigens, Tregs could play a major role in tumor progression. Accordingly, it has discovered that prostate cancer patients with higher Tregs have poor prognosis and low survival rates. However, anti-tumor responses can be reinforced by suppression of Tregs with using monoclonal antibodies against CD25 and CTLA-4. Therefore, depleting Tregs or suppressing their functions could be one of the effective ways for prostate cancer immunotherapy. The purpose of this review is to investigate the role of Treg cells in the progression of prostate cancer and to evaluate effective strategies for the treatment of prostate cancer by regulating Treg cells.
Collapse
|
11
|
Wengner AM, Scholz A, Haendler B. Targeting DNA Damage Response in Prostate and Breast Cancer. Int J Mol Sci 2020; 21:E8273. [PMID: 33158305 PMCID: PMC7663807 DOI: 10.3390/ijms21218273] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
Steroid hormone signaling induces vast gene expression programs which necessitate the local formation of transcription factories at regulatory regions and large-scale alterations of the genome architecture to allow communication among distantly related cis-acting regions. This involves major stress at the genomic DNA level. Transcriptionally active regions are generally instable and prone to breakage due to the torsional stress and local depletion of nucleosomes that make DNA more accessible to damaging agents. A dedicated DNA damage response (DDR) is therefore essential to maintain genome integrity at these exposed regions. The DDR is a complex network involving DNA damage sensor proteins, such as the poly(ADP-ribose) polymerase 1 (PARP-1), the DNA-dependent protein kinase catalytic subunit (DNA-PKcs), the ataxia-telangiectasia-mutated (ATM) kinase and the ATM and Rad3-related (ATR) kinase, as central regulators. The tight interplay between the DDR and steroid hormone receptors has been unraveled recently. Several DNA repair factors interact with the androgen and estrogen receptors and support their transcriptional functions. Conversely, both receptors directly control the expression of agents involved in the DDR. Impaired DDR is also exploited by tumors to acquire advantageous mutations. Cancer cells often harbor germline or somatic alterations in DDR genes, and their association with disease outcome and treatment response led to intensive efforts towards identifying selective inhibitors targeting the major players in this process. The PARP-1 inhibitors are now approved for ovarian, breast, and prostate cancer with specific genomic alterations. Additional DDR-targeting agents are being evaluated in clinical studies either as single agents or in combination with treatments eliciting DNA damage (e.g., radiation therapy, including targeted radiotherapy, and chemotherapy) or addressing targets involved in maintenance of genome integrity. Recent preclinical and clinical findings made in addressing DNA repair dysfunction in hormone-dependent and -independent prostate and breast tumors are presented. Importantly, the combination of anti-hormonal therapy with DDR inhibition or with radiation has the potential to enhance efficacy but still needs further investigation.
Collapse
Affiliation(s)
| | | | - Bernard Haendler
- Preclinical Research, Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany; (A.M.W.); (A.S.)
| |
Collapse
|
12
|
Burdak-Rothkamm S, Mansour WY, Rothkamm K. DNA Damage Repair Deficiency in Prostate Cancer. Trends Cancer 2020; 6:974-984. [DOI: 10.1016/j.trecan.2020.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022]
|
13
|
Devos G, Berghen C, Van Eecke H, Stichele AV, Van Poppel H, Goffin K, Mai C, De Wever L, Albersen M, Everaerts W, De Meerleer G, Joniau S. Oncological Outcomes of Metastasis-Directed Therapy in Oligorecurrent Prostate Cancer Patients Following Radical Prostatectomy. Cancers (Basel) 2020; 12:E2271. [PMID: 32823690 PMCID: PMC7464259 DOI: 10.3390/cancers12082271] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/25/2022] Open
Abstract
Several retrospective and a few prospective studies have shown that metastasis-directed therapy (MDT) could delay clinical progression and postpone the initiation of systemic treatment in oligorecurrent prostate cancer (PCa) patients. However, these endpoints are strongly influenced by variables such as concomitant use of androgen deprivation therapy (ADT) and follow-up imaging protocols. The aim of this manuscript was to assess palliative ADT- and metastatic castration-resistant prostate cancer (mCRPC)-free survival as long-term oncological outcomes in oligorecurrent PCa treated by MDT. We retrospectively identified consecutive post-prostatectomy oligorecurrent PCa patients treated by MDT (salvage lymphadenectomy, radiotherapy, or metastasectomy) at our tertiary referral center. Patients were eligible for inclusion if they developed recurrence following radical prostatectomy, had ≤5 metastatic lesions on imaging and had a serum testosterone >50 ng/dL or a testosterone suppression therapy-free interval of >2 years prior to the first MDT as an assumption of recovered serum testosterone (if no testosterone measurement available). Patients with castration-resistant or synchronous oligometastatic PCa at the time of first MDT were excluded. Repeated MDTs were allowed, as well as a period of concomitant ADT. Kaplan-Meier analyses were performed to assess palliative ADT-free and mCRPC-free survival. We identified 191 eligible patients who underwent MDT. Median follow-up from first MDT until last follow-up or death was 45 months (IQR 27-70; mean 51 months). Estimated median palliative-ADT free survival was 66 months (95% CI 58-164) and estimated median mCRPC-free survival was not reached (mean 117 months, 95% CI 103-132). In total, 314 MDTs were performed and 25 patients (13%) received ≥3 MDTs. This study demonstrated that (repeated) MDT is feasible and holds promise in terms of palliative ADT-free and mCRPC-free survival for patients with oligorecurrent PCa. However, these findings should be confirmed in prospective randomized controlled trials.
Collapse
Affiliation(s)
- Gaëtan Devos
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium; (H.V.E.); (A.V.S.); (H.V.P.); (M.A.); (W.E.)
| | - Charlien Berghen
- Department of Radiation Oncology, University Hospitals Leuven, Leuven 3000, Belgium; (C.B.); (G.D.M.)
| | - Henri Van Eecke
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium; (H.V.E.); (A.V.S.); (H.V.P.); (M.A.); (W.E.)
| | - Arthur Vander Stichele
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium; (H.V.E.); (A.V.S.); (H.V.P.); (M.A.); (W.E.)
| | - Hendrik Van Poppel
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium; (H.V.E.); (A.V.S.); (H.V.P.); (M.A.); (W.E.)
| | - Karolien Goffin
- Department of Nuclear Medicine, University Hospitals Leuven, Leuven 3000, Belgium;
| | - Cindy Mai
- Department of Radiology, University Hospitals Leuven, Leuven 3000, Belgium; (L.D.W.); (C.M.)
| | - Liesbeth De Wever
- Department of Radiology, University Hospitals Leuven, Leuven 3000, Belgium; (L.D.W.); (C.M.)
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium; (H.V.E.); (A.V.S.); (H.V.P.); (M.A.); (W.E.)
| | - Wouter Everaerts
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium; (H.V.E.); (A.V.S.); (H.V.P.); (M.A.); (W.E.)
| | - Gert De Meerleer
- Department of Radiation Oncology, University Hospitals Leuven, Leuven 3000, Belgium; (C.B.); (G.D.M.)
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium; (H.V.E.); (A.V.S.); (H.V.P.); (M.A.); (W.E.)
| |
Collapse
|
14
|
Philippou Y, Sjoberg H, Lamb AD, Camilleri P, Bryant RJ. Harnessing the potential of multimodal radiotherapy in prostate cancer. Nat Rev Urol 2020; 17:321-338. [PMID: 32358562 DOI: 10.1038/s41585-020-0310-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
Radiotherapy in combination with androgen deprivation therapy (ADT) is a standard treatment option for men with localized and locally advanced prostate cancer. However, emerging clinical evidence suggests that radiotherapy can be incorporated into multimodality therapy regimens beyond ADT, in combinations that include chemotherapy, radiosensitizing agents, immunotherapy and surgery for the treatment of men with localized and locally advanced prostate cancer, and those with oligometastatic disease, in whom the low metastatic burden in particular might be treatable with these combinations. This multimodal approach is increasingly recognized as offering considerable clinical benefit, such as increased antitumour effects and improved survival. Thus, radiotherapy is becoming a key component of multimodal therapy for many stages of prostate cancer, particularly oligometastatic disease.
Collapse
Affiliation(s)
- Yiannis Philippou
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Hanna Sjoberg
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Philip Camilleri
- Oxford Department of Clinical Oncology, Churchill Hospital Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Headington, Oxford, UK
| | - Richard J Bryant
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK.
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK.
| |
Collapse
|
15
|
[Indications and outlooks of radiohormonal therapy of high-risk prostate cancers]. Cancer Radiother 2020; 24:143-152. [PMID: 32057646 DOI: 10.1016/j.canrad.2019.06.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/21/2019] [Accepted: 06/19/2019] [Indexed: 11/21/2022]
Abstract
Prostate cancer is a sensitive adenocarcinoma, in more than 80% of cases, to chemical castration, due to its hormone dependence. Locally advanced and/or high-risk cancer is defined based on clinical stage, initial prostate specific antigen serum concentration value or high Gleason score. Hormone therapy associated with radiation therapy is the standard of management and improves local control, reduces the risk of distant metastasis and improves specific and overall survival. Duration of hormone therapy, dose level of radiation therapy alone or associated with brachytherapy are controversial data in the literature. The therapeutic choice, multidisciplinary, depends on the age and comorbidity of the patient, the prognostic criteria of the pathology and the urinary function of the patient. Current research focuses on optimizing local and distant control of these aggressive forms and incorporates neoadjuvant or adjuvant chemotherapy and also new hormone therapies.
Collapse
|
16
|
Latorzeff I. [Optimizing local control of high-risk prostate cancers through multimodal treatments]. Prog Urol 2019; 29 Suppl 1:S8-S19. [PMID: 31307631 DOI: 10.1016/s1166-7087(19)30166-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prostate cancer is a sensitive adenocarcinoma, in more than 80 % of cases, to chemical castration, due to its hormone dependence. Locally advanced and/or high-risk cancer is defined based on clinical stage, initial PSA value or high Gleason score. Hormone therapy associated with radiation therapy is the standard of management and improves local control, reduces the risk of distant metastasis and improves specific and overall survival. Duration of hormone therapy, dose level of radiation therapy alone or associated with brachytherapy are controversial data in the literature. Radical prostatectomy surgery is a therapeutic option that must be performed with extensive lymph node dissection and is often part of a multimodal care sequence. The therapeutic choice, multidisciplinary, depends on the age and co-morbidity of the patient, the prognostic criteria of the pathology and the urinary function of the patient. Current research focuses on optimizing local and distant control of these aggressive forms and incorporates neo-adjuvant or adjuvant chemotherapy and also new hormone therapies.
Collapse
Affiliation(s)
- I Latorzeff
- Oncologie-radiothérapie, Bât Atrium, clinique Pasteur, 1, rue de la Petite-Vitesse, 31300 Toulouse, France.
| |
Collapse
|
17
|
Arcangeli S, Jereczek-Fossa BA, Alongi F, Aristei C, Becherini C, Belgioia L, Buglione M, Caravatta L, D'Angelillo RM, Filippi AR, Fiore M, Genovesi D, Greco C, Livi L, Magrini SM, Marvaso G, Mazzola R, Meattini I, Merlotti A, Palumbo I, Pergolizzi S, Ramella S, Ricardi U, Russi E, Trovò M, Sindoni A, Valentini V, Corvò R. Combination of novel systemic agents and radiotherapy for solid tumors - part I: An AIRO (Italian association of radiotherapy and clinical oncology) overview focused on treatment efficacy. Crit Rev Oncol Hematol 2018; 134:87-103. [PMID: 30658886 DOI: 10.1016/j.critrevonc.2018.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Over the past century, technologic advances have promoted the evolution of radiation therapy into a precise treatment modality allowing for the maximal administration of dose to tumors while sparing normal tissues. In parallel with this technological maturation, the rapid expansion in understanding the basic biology and heterogeneity of cancer has led to the development of several compounds that target specific pathways. Many of them are in advanced steps of clinical development for combination treatments with radiotherapy, and can be incorporated into radiation oncology practice for a personalized approach to maximize the therapeutic gain. This review describes the rationale for combining novel agents with radiation, and provides an overview of the current landscape focused on treatment efficacy.
Collapse
Affiliation(s)
- Stefano Arcangeli
- Department of Radiation Oncology, Policlinico S. Gerardo and University of Milan "Bicocca", Milan, Italy.
| | | | - Filippo Alongi
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, and University of Brescia, Brescia, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Carlotta Becherini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Liliana Belgioia
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| | - Michela Buglione
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Luciana Caravatta
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | | | | | - Michele Fiore
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Domenico Genovesi
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | - Carlo Greco
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Stefano Maria Magrini
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Giulia Marvaso
- Deparment of Radiation Oncology of IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Rosario Mazzola
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, and University of Brescia, Brescia, Italy
| | - Icro Meattini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Anna Merlotti
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Isabella Palumbo
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Stefano Pergolizzi
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Italy
| | - Sara Ramella
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | | | - Elvio Russi
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Marco Trovò
- Department of Radiation Oncology, Azienda Sanitaria Universitaria Integrata of Udine, Udine, Italy
| | - Alessandro Sindoni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Valentini
- Gemelli Advanced Radiation Therapy Center, Fondazione Policlinico Universitario "A. Gemelli", Catholic University of Sacred Heart, Rome, Italy
| | - Renzo Corvò
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| |
Collapse
|
18
|
Tosco L, Briganti A, D'amico AV, Eastham J, Eisenberger M, Gleave M, Haustermans K, Logothetis CJ, Saad F, Sweeney C, Taplin ME, Fizazi K. Systematic Review of Systemic Therapies and Therapeutic Combinations with Local Treatments for High-risk Localized Prostate Cancer. Eur Urol 2018; 75:44-60. [PMID: 30286948 DOI: 10.1016/j.eururo.2018.07.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/17/2018] [Indexed: 01/09/2023]
Abstract
CONTEXT Systemic therapies, combined with local treatment for high-risk prostate cancer, are recommended by the international guidelines for specific subgroups of patients; however, for many of the clinical scenarios, it remains a research field. OBJECTIVE To perform a systematic review, and describe current evidence and perspectives about the multimodal treatment of high-risk prostate cancer. EVIDENCE ACQUISITION We performed a systematic review of PubMED, Embase, Cochrane Library, European Society of Medical Oncology/American Society of Clinical Oncology Annual proceedings, and clinicalTrial.gov between January 2010 and February 2018 following the Preferred Reporting Items for Systematic Reviews and Meta-analysis statement. EVIDENCE SYNTHESIS Seventy-seven prospective trials were identified. According to multiple randomized trials, combining androgen deprivation therapy (ADT) with external-beam radiotherapy (EBRT) outperforms EBRT alone for both relapse-free and overall survival. Neoadjuvant ADT did not show significant improvement compared with prostatectomy alone. The role of adjuvant ADT after prostatectomy in patients with high-risk disease is still debated, with lack of data from phase 3 trials in pN0 patients. Novel androgen pathway inhibitors have been tested only in early-phase trials in addition to primary treatment. GETUG 12, RTOG 0521, and nonmetastatic subgroup of the STAMPEDE trial showed improved relapse-free survival for docetaxel in patients treated with EBRT plus ADT, although mature metastasis-free survival data are still pending. Both the SPCG-12 and the VACSP#553 trial showed no improvement in relapse-free survival for adjuvant docetaxel after prostatectomy. CONCLUSIONS In contrast to the clearly demonstrated survival benefits of long-term adjuvant ADT when used with EBRT, its role after prostatectomy remains unclear especially in pN0 patients. Adding docetaxel to EBRT-ADT improves relapse-free survival, with immature results on overall survival. Novel androgen receptor pathway inhibitors are currently being tested in the neoadjuvant and adjuvant setting. PATIENT SUMMARY Treatment of high-risk prostate cancer is based on a multimodality approach that includes systemic treatments. The best treatment or therapy combination remains to be defined.
Collapse
Affiliation(s)
- Lorenzo Tosco
- Department of Urology, University Hospitals Leuven, Leuven, Belgium; Nuclear Medicine & Molecular Imaging, KU Leuven, Leuven, Belgium.
| | - Alberto Briganti
- Division of Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Antony Vincent D'amico
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - James Eastham
- Urology Service at the Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mario Eisenberger
- Department of Oncology in the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Martin Gleave
- The Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Karin Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fred Saad
- Department of Urology, Centre Hospitalier de l'Université de Montréal, University of Montreal, Montreal, Quebec, Canada
| | | | | | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Sud, Villejuif, France
| |
Collapse
|
19
|
Sensitization of prostate cancer to radiation therapy: Molecules and pathways to target. Radiother Oncol 2018; 128:283-300. [PMID: 29929859 DOI: 10.1016/j.radonc.2018.05.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/01/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022]
Abstract
Radiation therapy is used to treat cancer by radiation-induced DNA damage. Despite the best efforts to eliminate cancer, some cancer cells survive irradiation, resulting in cancer progression or recurrence. Alteration in DNA damage repair pathways is common in cancers, resulting in modulation of their response to radiation. This article focuses on the recent findings about molecules and pathways that potentially can be targeted to sensitize prostate cancer cells to ionizing radiation, thereby achieving an improved therapeutic outcome.
Collapse
|
20
|
Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 2018; 19:975-986. [PMID: 29880291 DOI: 10.1016/s1470-2045(18)30365-6] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with metastatic castration-resistant prostate cancer and homologous recombination repair (HRR) mutations have a better response to treatment with the poly(ADP-ribose) polymerase inhibitor olaparib than patients without HRR mutations. Preclinical data suggest synergy between olaparib and androgen pathway inhibitors. We aimed to assess the efficacy of olaparib plus the androgen pathway inhibitor abiraterone in patients with metastatic castration-resistant prostate cancer regardless of HRR mutation status. METHODS We carried out this double-blind, randomised, placebo-controlled phase 2 trial at 41 urological oncology sites in 11 countries across Europe and North America. Eligible male patients were aged 18 years or older with metastatic castration-resistant prostate cancer who had previously received docetaxel and were candidates for abiraterone treatment. Patients were excluded if they had received more than two previous lines of chemotherapy, or had previous exposure to second-generation antihormonal drugs. Patients were randomly assigned (1:1) using an interactive voice or web response system, without stratification, to receive oral olaparib 300 mg twice daily or placebo. All patients received oral abiraterone 1000 mg once daily and prednisone or prednisolone 5 mg twice daily. Patients and investigators were masked to treatment allocation. The primary endpoint was investigator-assessed radiographic progression-free survival (rPFS; based on Response Evaluation Criteria in Solid Tumors version 1.1 and Prostate Cancer Clinical Trials Working Group 2 criteria). Efficacy analyses were done in the intention-to-treat population, which included all randomly assigned patients, and safety analyses included all patients who received at least one dose of olaparib or placebo. This trial is registered with ClinicalTrials.gov, number NCT01972217, and is no longer recruiting patients. FINDINGS Between Nov 25, 2014, and July 14, 2015, 171 patients were assessed for eligibility. Of those, 142 patients were randomly assigned to receive olaparib and abiraterone (n=71) or placebo and abiraterone (n=71). The clinical cutoff date for the final analysis was Sept 22, 2017. Median rPFS was 13·8 months (95% CI 10·8-20·4) with olaparib and abiraterone and 8·2 months (5·5-9·7) with placebo and abiraterone (hazard ratio [HR] 0·65, 95% CI 0·44-0·97, p=0·034). The most common grade 1-2 adverse events were nausea (26 [37%] patients in the olaparib group vs 13 [18%] patients in the placebo group), constipation (18 [25%] vs eight [11%]), and back pain (17 [24%] vs 13 [18%]). 38 (54%) of 71 patients in the olaparib and abiraterone group and 20 (28%) of 71 patients in the placebo and abiraterone group had grade 3 or worse adverse events, including anaemia (in 15 [21%] of 71 patients vs none of 71), pneumonia (four [6%] vs three [4%]), and myocardial infarction (four [6%] vs none). Serious adverse events were reported by 24 (34%) of 71 patients receiving olaparib and abiraterone (seven of which were related to treatment) and 13 (18%) of 71 patients receiving placebo and abiraterone (one of which was related to treatment). One treatment-related death (pneumonitis) occurred in the olaparib and abiraterone group. INTERPRETATION Olaparib in combination with abiraterone provided clinical efficacy benefit for patients with metastatic castration-resistant prostate cancer compared with abiraterone alone. More serious adverse events were observed in patients who received olaparib and abiraterone than abiraterone alone. Our data suggest that the combination of olaparib and abiraterone might provide an additional clinical benefit to a broad population of patients with metastatic castration-resistant prostate cancer. FUNDING AstraZeneca.
Collapse
|
21
|
Ide H, Inoue S, Mizushima T, Jiang G, Chuang KH, Oya M, Miyamoto H. Androgen Receptor Signaling Reduces Radiosensitivity in Bladder Cancer. Mol Cancer Ther 2018; 17:1566-1574. [PMID: 29720561 DOI: 10.1158/1535-7163.mct-17-1061] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/28/2017] [Accepted: 04/27/2018] [Indexed: 11/16/2022]
Abstract
Although radiotherapy often with chemotherapy has been shown to offer a survival benefit comparable with that of radical cystectomy in select patients with bladder cancer, the development of radiosensitization strategies may significantly enhance its application. Notably, emerging preclinical evidence has indicated the involvement of androgen receptor (AR) signaling in urothelial cancer progression. We here assessed whether AR signals could contribute to modulating radiosensitivity in bladder cancer cells. Ionizing radiation reduced the numbers of viable cells or colonies of AR-negative lines more significantly than those of AR-positive lines. Similarly, in AR-positive cells cultured in androgen-depleted conditions, dihydrotestosterone treatment lowered the effects of irradiation. Meanwhile, an antiandrogen hydroxyflutamide enhanced them in AR-positive cells cultured in the presence of androgens. AR knockdown or hydroxyflutamide treatment also resulted in a delay in DNA double-strand break repair 4-24 hours after irradiation. We then established "radiation-resistant" sublines and found considerable elevation of the expression of AR as well as DNA repair genes, such as ATR, CHEK1, and PARP-1, in these sublines, compared with respective controls. Furthermore, dihydrotestosterone induced the expression of these DNA repair genes in irradiated AR-positive cells, and hydroxyflutamide antagonized the androgen effects. Finally, in a mouse xenograft model, low-dose flutamide was found to enhance the inhibitory effects of irradiation, and its tumor size was similar to that of AR knockdown line with radiation alone. These findings suggest that AR activity inversely correlates with radiosensitivity in bladder cancer. Accordingly, antiandrogenic drugs may function as sensitizers of irradiation, especially in patients with AR-positive urothelial cancer. Mol Cancer Ther; 17(7); 1566-74. ©2018 AACR.
Collapse
Affiliation(s)
- Hiroki Ide
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Inoue
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, New York.,James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Taichi Mizushima
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, New York.,James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Guiyang Jiang
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, New York.,James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Kuang-Hsiang Chuang
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Miyamoto
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, New York.,James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York.,Department of Urology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
22
|
Tosco L, Laenen A, Gevaert T, Salmon I, Decaestecker C, Davicioni E, Buerki C, Claessens F, Swinnen J, Goffin K, Oyen R, Everaerts W, Moris L, De Meerleer G, Haustermans K, Joniau S. Neoadjuvant degarelix with or without apalutamide followed by radical prostatectomy for intermediate and high-risk prostate cancer: ARNEO, a randomized, double blind, placebo-controlled trial. BMC Cancer 2018; 18:354. [PMID: 29606109 PMCID: PMC5879743 DOI: 10.1186/s12885-018-4275-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 03/21/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Recent retrospective data suggest that neoadjuvant androgen deprivation therapy can improve the prognosis of high-risk prostate cancer (PCa) patients. Novel androgen receptor pathway inhibitors are nowadays available for treatment of metastatic PCa and these compounds are promising for early stage disease. Apalutamide is a pure androgen antagonist with a very high affinity with the androgen receptor. The combination of apalutamide with degarelix, an LHRH antagonist, could increase the efficacy compared to degarelix alone. OBJECTIVE The primary objective is to assess the difference in proportions of minimal residual disease at prostatectomy specimen between apalutamide + degarelix vs placebo + degarelix. Various secondary endpoints are assessed: variations of different biomarkers at the tumour level (tissue microarrays to evaluate DNA-PKs, PARP, AR and splice variants, PSMA, etc.), whole transcriptome sequencing, exome sequencing and clinical (PSA and testosterone kinetics, early biochemical recurrence free survival, quality of life, safety, etc.) and radiological endpoints. METHODS ARNEO is a single centre, phase II, randomized, double blind, placebo-controlled trial. The plan is to include at least 42 patients per each of the two study arms. Patients with intermediate/high-risk PCa and who are amenable for radical prostatectomy with pelvic lymph node dissection can be included. After signing an informed consent, every patient will undergo a pelvic 68Ga -PSMA-11 PSMA PET/MR and receive degarelix at standard dosage and start assuming apalutamide/placebo (60 mg 4 tablets/day) for 12 weeks. Within thirty days from the last study medication intake the same imaging will be repeated. Every patient will undergo PSA and testosterone testing the day of randomization, before the first drug intake, and after the last dose. Formalin fixed paraffin embedded tumour samples will be collected and used for transcriptome analysis, exome sequencing and immunohistochemistry. DISCUSSION ARNEO will allow us to answer, first, whether the combined treatment can result in an increased proportion of patients with minimal residual disease. Secondly, It will enable the study of the molecular consequences at the level of the tumour. Thirdly, what the consequences are of new generation androgen receptor pathway inhibitors on 68Ga -PSMA-11 PET/MR. Finally, various clinical, safety and quality of life data will be collected. TRIAL REGISTRATION EUDRaCT number: 2016-002854-19 (authorization date 3rd August 2017). clinicalTrial.gov: NCT03080116 .
Collapse
Affiliation(s)
- Lorenzo Tosco
- Urology, Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Annouschka Laenen
- Leuven Biostatistics and Statistical Bioinformatics Center, KU Leuven, Leuven, Belgium
| | - Thomas Gevaert
- Laboratory of Experimental Urology, Organ Systems, KU Leuven, Leuven, Belgium
- Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Department of Pathology, AZ Klina, Brasschaat, Belgium
| | - Isabelle Salmon
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Decaestecker
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratories of Image, Signal processing & Acoustics, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | - Frank Claessens
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, Leuven, Belgium
| | - Johan Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
- Leuven Cancer Institute, KU Leuven, University of Leuven, Leuven, Belgium
| | - Karolien Goffin
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, UZ Leuven, Leuven, Belgium
| | - Raymond Oyen
- Department of Radiology Gasthuisberg University Hospitals Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Urology, Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | - Lisa Moris
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, Leuven, Belgium
| | - Gert De Meerleer
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Karin Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Steven Joniau
- Urology, Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
23
|
Suppression of PC-1/PrLZ sensitizes prostate cancer cells to ionizing radiation by attenuating DNA damage repair and inducing autophagic cell death. Oncotarget 2018; 7:62340-62351. [PMID: 27694690 PMCID: PMC5308731 DOI: 10.18632/oncotarget.11470] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 08/09/2016] [Indexed: 01/18/2023] Open
Abstract
Radiotherapy is promising and effective for treating prostate cancer but the addition of a tumor cell radiosensitizer would improve therapeutic outcomes. PC-1/PrLZ, a TPD52 protein family member is frequently upregulated in advanced prostate cancer cells and may be a biomarker of aggressive prostate cancer. Therefore, we investigated the potential role of PC-1/PrLZ for increasing radioresistance in human prostate cancer cell lines. Growth curves and survival assays after g-ray irradiation confirmed that depletion of endogenous PC-1/PrLZ significantly increased prostate cancer cell radiosensitivity. Irradiation (IR) increased PC-1/PrLZ expression in a dose- and time-dependent manner and increased radiosensitivity in PC-1/PrLZ-suppressed cells was partially due to decreased DNA double strand break (DBS) repair which was measured with comet and gH2AX foci assays. Furthermore, depletion of PC-1/PrLZ impaired the IR-induced G2/M checkpoint, which has been reported to be correlate with radioresistance in cancer cells. PC-1/PrLZ-deficient cells exhibited higher level of autophagy when compared with control cells. Thus, specific inhibition of PC-1/PrLZ might provide a novel therapeutic strategy for radiosensitizing prostate cancer cells.
Collapse
|
24
|
Srivastava P, Sarma A, Chaturvedi CM. Targeting DNA repair with PNKP inhibition sensitizes radioresistant prostate cancer cells to high LET radiation. PLoS One 2018; 13:e0190516. [PMID: 29320576 PMCID: PMC5762163 DOI: 10.1371/journal.pone.0190516] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/15/2017] [Indexed: 12/19/2022] Open
Abstract
High linear energy transfer (LET) radiation or heavy ion such as carbon ion radiation is used as a method for advanced radiotherapy in the treatment of cancer. It has many advantages over the conventional photon based radiotherapy using Co-60 gamma or high energy X-rays from a Linear Accelerator. However, charged particle therapy is very costly. One way to reduce the cost as well as irradiation effects on normal cells is to reduce the dose of radiation by enhancing the radiation sensitivity through the use of a radiomodulator. PNKP (polynucleotide kinase/phosphatase) is an enzyme which plays important role in the non-homologous end joining (NHEJ) DNA repair pathway. It is expected that inhibition of PNKP activity may enhance the efficacy of the charged particle irradiation in the radioresistant prostate cancer cell line PC-3. To test this hypothesis, we investigated cellular radiosensitivity by clonogenic cell survival assay in PC-3 cells.12Carbon ion beam of62 MeVenergy (equivalent 5.16 MeV/nucleon) and with an entrance LET of 287 kev/μm was used for the present study. Apoptotic parameters such as nuclear fragmentation and caspase-3 activity were measured by DAPI staining, nuclear ladder assay and colorimetric caspase-3method. Cell cycle arrest was determined by FACS analysis. Cell death was enhanced when carbon ion irradiation is combined with PNKPi (PNKP inhibitor) to treat cells as compared to that seen for PNKPi untreated cells. A low concentration (10μM) of PNKPi effectively radiosensitized the PC-3 cells in terms of reduction of dose in achieving the same survival fraction. PC-3 cells underwent significant apoptosis and cell cycle arrest too was enhanced at G2/M phase when carbon ion irradiation was combined with PNKPi treatment. Our findings suggest that combined treatment of carbon ion irradiation and PNKP inhibition could enhance cellular radiosensitivity in a radioresistant prostate cancer cell line PC-3. The synergistic effect of PNKPi and carbon ion irradiation could be used as a promising method for carbon-ion therapy in radioresistant cells.
Collapse
Affiliation(s)
- Pallavi Srivastava
- Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Asitikantha Sarma
- Radiation Biology Laboratory, Inter University Accelerator Centre, New Delhi, India
| | | |
Collapse
|
25
|
Ghashghaei M, Niazi TM, Heravi M, Bekerat H, Trifiro M, Paliouras M, Muanza T. Enhanced radiosensitization of enzalutamide via schedule dependent administration to androgen-sensitive prostate cancer cells. Prostate 2018; 78:64-75. [PMID: 29134684 DOI: 10.1002/pros.23445] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 10/10/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Prostate cancer (PCa) is a progressive disease and the most diagnosed cancer in men. The current standard of care for high-risk localized PCa is a combination of androgen deprivation therapy (ADT) and radiation (XRT). The majority of these patients however become resistant due to incomplete responses to ADT as a result of selective cells maintaining androgen receptor (AR) activity. Improvement can be made if increasing radiosensitivity is realized. Therefore, the aim of this study is to investigate the efficacy of the next-generation PCa drug Enzalutamide (ENZA), as a radiosensitizer in XRT therapy. METHODS Using a number of androgen-dependent (LNCaP, PC3-T877A) and androgen-independent (C4-2, 22RV1, PC3, PC3-AR V7) cell lines, the effect of ENZA as a radiosensitizer was studied alone or in combination with ADT and/or XRT. Cell viability and cell survival were assessed, along with determination of cell cycle arrest, DNA damage response and repair, apoptosis and senescence. RESULTS Our results indicated that either ENZA alone (in AR positive, androgen-dependent PCa cells) or in combination with ADT (in AR positive, hormone-insensitive PCa cells) potentiates radiation response [Dose enhancement factor (DEF) of 1.75 in LNCAP and 1.35 in C4-2] stronger than ADT + XRT conditions. Additionally, ENZA sensitized androgen dependent PCa cells to XRT in a schedule-dependent manner, where concurrent administration of ENZA and radiation lead to a maximal radiosensitization when compared to either drug administration prior or after XRT. In LNCaP cells, ENZA treatment significantly prolonged the presence of XRT-induced phospho-γH2AX up to 24 h after treatment; suggesting enhanced DNA damage. It also significantly increased XRT-induced apoptosis and senescence. CONCLUSIONS Our data indicates that ENZA acts as a much stronger radiosensitizer compared to ADT. We have also observed that its efficacy is schedule dependent and related to increased levels of DNA damage and a delay of DNA repair processes. Finally, the initial abrogation of DNA-PKcs activity by AR inhibition and its subsequent recovery might represent an important mechanism by which PCa cells acquire resistance to combined anti-androgen and XRT treatment. This work suggests a new use of ENZA in combination with XRT that could be applicable in clinical trial settings for patients with early and intermediate hormone responsive disease.
Collapse
Affiliation(s)
- Maryam Ghashghaei
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Tamim M Niazi
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Department of Radiation Oncology, Jewish General Hospital, Quebec, Canada
| | - Mitra Heravi
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Hamed Bekerat
- Department of Radiation Oncology, Jewish General Hospital, Quebec, Canada
| | - Mark Trifiro
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Endocrinology, Jewish General Hospital, Montreal, Quebec, Canada
| | - Miltiadis Paliouras
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Thierry Muanza
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Department of Radiation Oncology, Jewish General Hospital, Quebec, Canada
| |
Collapse
|
26
|
Hennequin C, Fumagalli I, Martin V, Quero L. [Combination of radiotherapy and androgen deprivation therapy for localized prostate cancer]. Cancer Radiother 2017; 21:462-468. [PMID: 28870416 DOI: 10.1016/j.canrad.2017.07.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/02/2017] [Indexed: 10/18/2022]
Abstract
Combination of radiotherapy and androgen deprivation is now considered as the standard of care for patients with a localized prostate cancer but poor prognosis factors. Two groups of randomized trials have led to this recommendation: some have compared radiotherapy alone versus hormonal treatment and radiotherapy: these trials demonstrated, now with a long follow-up, an improvement in 10-year survival for the combined treatment. Three recent trials compared androgen deprivation alone or combined with radiotherapy; a benefit in survival was also demonstrated in favour of the combination. Some questions remained concerning the optimal duration of hormonal treatment, in view of its potential side effects. Patients in the intermediate prognostic groups could receive a short-term androgen deprivation, but those with a high Gleason score must be treated with a long-term hormonal treatment. Modalities of radiotherapy, regarding volumes and dose must also be précised in the next years.
Collapse
Affiliation(s)
- C Hennequin
- Service de cancérologie-radiothérapie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75475 Paris, France.
| | - I Fumagalli
- Service de cancérologie-radiothérapie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75475 Paris, France
| | - V Martin
- Service de cancérologie-radiothérapie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75475 Paris, France
| | - L Quero
- Service de cancérologie-radiothérapie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, 75475 Paris, France
| |
Collapse
|
27
|
Tosco L, Laenen A, Briganti A, Gontero P, Karnes RJ, Albersen M, Bastian PJ, Chlosta P, Claessens F, Chun FK, Everaerts W, Gratzke C, Graefen M, Kneitz B, Marchioro G, Salas RS, Tombal B, Van den Broeck T, Moris L, Battaglia A, van der Poel H, Walz J, Bossi A, De Meerleer G, Haustermans K, Van Poppel H, Spahn M, Joniau S. The survival impact of neoadjuvant hormonal therapy before radical prostatectomy for treatment of high-risk prostate cancer. Prostate Cancer Prostatic Dis 2017; 20:407-412. [PMID: 28485390 DOI: 10.1038/pcan.2017.29] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 04/07/2017] [Accepted: 04/08/2017] [Indexed: 11/09/2022]
Abstract
BACKGROUND Several randomized controlled trials assessed the outcomes of patients treated with neoadjuvant hormonal therapy (NHT) before radical prostatectomy (RP). The majority of them included mainly low and intermediate risk prostate cancer (PCa) without specifically assessing PCa-related death (PCRD). Thus, there is a lack of knowledge regarding a possible effect of NHT on PCRD in the high-risk PCa population. We aimed to analyze the effect of NHT on PCRD in a multicenter high-risk PCa population treated with RP, using a propensity-score adjustment. METHODS This is a retrospective multi-institutional study including patients with high-risk PCa defined as: clinical stage T3-4, PSA >20 ng ml-1 or biopsy Gleason score 8-10. We compared PCRD between RP and NHT+RP using competing risks analysis. Correction for group differences was performed by propensity-score adjustment. RESULTS After application of the inclusion/exclusion criteria, 1573 patients remained for analysis; 1170 patients received RP and 403 NHT+RP. Median follow-up was 56 months (interquartile range 29-88). Eighty-six patients died of PCa and 106 of other causes. NHT decreased the risk of PCRD (hazard ratio (HR) 0.5; 95% confidence interval (CI) 0.32-0.80; P=0.0014). An interaction effect between NHT and radiotherapy (RT) was observed (HR 0.3; 95% CI 0.21-0.43; P<0.0008). More specifically, of patients who received adjuvant RT, those who underwent NHT+RP had decreased PCRD rates (2.3% at 5 year) compared to RP (7.5% at 5 year). The retrospective design and lack of specific information about NHT are possible limitations. CONCLUSIONS In this propensity-score adjusted analysis from a large high-risk PCa population, NHT before surgery significantly decreased PCRD. This effect appeared to be mainly driven by the early addition of RT post-surgery. The specific sequence of NHT+RP and adjuvant RT merits further study in the high-risk PCa population.
Collapse
Affiliation(s)
- L Tosco
- Department of Development and Regeneration, University Hospitals Leuven, Urology, Leuven, Belgium.,Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
| | - A Laenen
- Department of Public Health and Primary Care, Leuven Biostatistics and Statistical Bioinformatics Center, KU Leuven, Leuven, Belgium
| | - A Briganti
- Department of Urology, San Raffaele Hospital, University VitaSalute, Milan, Italy
| | - P Gontero
- Department of University Urology, Urologia U, Città della Salue e della Scienza di Torino, Molinette Hospital, Turin, Italy
| | - R J Karnes
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - M Albersen
- Department of Development and Regeneration, University Hospitals Leuven, Urology, Leuven, Belgium
| | - P J Bastian
- Department of Urology, Urologische Klinik Und Poliklinik, Klinikum Der Universität München Campus Großhadern, Munich, Germany
| | - P Chlosta
- Department of Urology, Jagiellonian University Medical College, Krakow, Poland
| | - F Claessens
- Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - F K Chun
- Department of Urology, University of Hamburg, Hamburg, Germany
| | - W Everaerts
- Department of Development and Regeneration, University Hospitals Leuven, Urology, Leuven, Belgium
| | - C Gratzke
- Department of Urology, Urologische Klinik Und Poliklinik, Klinikum Der Universität München Campus Großhadern, Munich, Germany
| | - M Graefen
- Department of Urology, Martini Klinik am UKE GmbH, Hamburg, Germany
| | - B Kneitz
- Department of Urology and Pediatric Urology, University Hospital Wurzburg, Wurzburg, Germany
| | - G Marchioro
- Department of Urology, University of Piemonte Orientale, Novara, Italy
| | - R S Salas
- Department of Urology, Institut Mutualiste Montsouris and Paris Descartes University, Paris, France
| | - B Tombal
- Department of Urology, Cliniques Universitaires SaintLuc, Brussels, Belgium
| | - T Van den Broeck
- Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - L Moris
- Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - A Battaglia
- Department of University Urology, Urologia U, Città della Salue e della Scienza di Torino, Molinette Hospital, Turin, Italy
| | - H van der Poel
- Department Of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J Walz
- Department of Urology, Institut Paoli Calmettes Cancer Centre, Marseille, France
| | - A Bossi
- Department of Radiation Oncology, Gustave Roussy Cancer Institute, Villejuif, France
| | - G De Meerleer
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - K Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - H Van Poppel
- Department of Development and Regeneration, University Hospitals Leuven, Urology, Leuven, Belgium
| | - M Spahn
- Department of Urology, University Hospital Bern, Inselspital, Berne, Switzerland
| | - S Joniau
- Department of Development and Regeneration, University Hospitals Leuven, Urology, Leuven, Belgium
| |
Collapse
|
28
|
Folate-targeted nanoparticle delivery of androgen receptor shRNA enhances the sensitivity of hormone-independent prostate cancer to radiotherapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1309-1321. [PMID: 28185938 DOI: 10.1016/j.nano.2017.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/21/2017] [Accepted: 01/28/2017] [Indexed: 12/15/2022]
Abstract
Androgen receptor (AR) plays a crucial role in the development and progression of prostate cancer (PCa). PCa patients typically receive androgen deprivation therapy; nonetheless, these patients eventually develop castration and radiation resistance. We hypothesized that we could further improve radiotherapeutic efficacy of hormone-independent PCa (HIPC) by silencing AR. In this study, nanoparticle (NP) AR-shRNA was formulated using folate-targeted H1 nanopolymer. We demonstrated that NP AR-shRNA enhances PCa radiosensitivity as indicated by the inhibition of cell growth, increased apoptosis, and increased cell cycle arrest in AR-dependent HIPC in vitro. The radiosensitizing effect of NP AR-shRNA could be validated in vivo, as NP AR-shRNA significantly suppressed tumor growth and prolonged the survival of HIPC tumor-bearing mice. Analysis at the molecular level revealed that NP AR-shRNA inhibits DNA damage repair signaling pathways. Our study supports further investigation of NP AR-shRNA for the improvement of radiotherapy efficacy in HIPC.
Collapse
|
29
|
Cellular Pathways in Response to Ionizing Radiation and Their Targetability for Tumor Radiosensitization. Int J Mol Sci 2016; 17:ijms17010102. [PMID: 26784176 PMCID: PMC4730344 DOI: 10.3390/ijms17010102] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/22/2015] [Accepted: 12/25/2015] [Indexed: 12/20/2022] Open
Abstract
During the last few decades, improvements in the planning and application of radiotherapy in combination with surgery and chemotherapy resulted in increased survival rates of tumor patients. However, the success of radiotherapy is impaired by two reasons: firstly, the radioresistance of tumor cells and, secondly, the radiation-induced damage of normal tissue cells located in the field of ionizing radiation. These limitations demand the development of drugs for either radiosensitization of tumor cells or radioprotection of normal tissue cells. In order to identify potential targets, a detailed understanding of the cellular pathways involved in radiation response is an absolute requirement. This review describes the most important pathways of radioresponse and several key target proteins for radiosensitization.
Collapse
|
30
|
Spratt DE, Evans MJ, Davis BJ, Doran MG, Lee MX, Shah N, Wongvipat J, Carnazza KE, Klee GG, Polkinghorn W, Tindall DJ, Lewis JS, Sawyers CL. Androgen Receptor Upregulation Mediates Radioresistance after Ionizing Radiation. Cancer Res 2015; 75:4688-96. [PMID: 26432404 DOI: 10.1158/0008-5472.can-15-0892] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 08/11/2015] [Indexed: 12/19/2022]
Abstract
Clinical trials have established the benefit of androgen deprivation therapy (ADT) combined with radiotherapy in prostate cancer. ADT sensitizes prostate cancer to radiotherapy-induced death at least in part through inhibition of DNA repair machinery, but for unknown reasons, adjuvant ADT provides further survival benefits. Here, we show that androgen receptor (AR) expression and activity are durably upregulated following radiotherapy in multiple human prostate cancer models in vitro and in vivo. Moreover, the degree of AR upregulation correlates with survival in vitro and time to tumor progression in animal models. We also provide evidence of AR pathway upregulation, measured by a rise in serum levels of AR-regulated hK2 protein, in nearly 20% of patients after radiotherapy. Furthermore, these men were three-fold more likely to experience subsequent biochemical failure. Collectively, these data demonstrate that radiotherapy can upregulate AR signaling after therapy to an extent that negatively affects disease progression and/or survival.
Collapse
Affiliation(s)
- Daniel E Spratt
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York. Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael J Evans
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian J Davis
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Michael G Doran
- Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Man Xia Lee
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neel Shah
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Wongvipat
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathryn E Carnazza
- Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - George G Klee
- Departments of Urology and Biochemistry/Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - William Polkinghorn
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York. Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Donald J Tindall
- Departments of Urology and Biochemistry/Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Jason S Lewis
- Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Charles L Sawyers
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
31
|
Gromov P, Espinoza JA, Gromova I. Molecular and diagnostic features of apocrine breast lesions. Expert Rev Mol Diagn 2015; 15:1011-22. [DOI: 10.1586/14737159.2015.1057125] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
32
|
Advances in prostate cancer research and treatment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:708383. [PMID: 25215290 PMCID: PMC4151599 DOI: 10.1155/2014/708383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 07/23/2014] [Indexed: 11/17/2022]
|