1
|
Dal Pra A, Ghadjar P, Ryu HM, Proudfoot JA, Hayoz S, Michalski JM, Spratt DE, Liu Y, Schär C, Berlin AM, Zwahlen DR, Simko JP, Hölscher T, Efstathiou JA, Polat B, Sandler HM, Hildebrandt G, Parliament MB, Mueller AC, Dayes IS, Plasswilm L, Correa RJM, Robertson JM, Karrison TG, Davicioni E, Hall WA, Feng FY, Pollack A, Thalmann GN, Nguyen PL, Aebersold DM, Tran PT, Zhao SG. Predicting dose response to prostate cancer radiotherapy: validation of a radiation signature in the randomized phase III NRG/RTOG 0126 and SAKK 09/10 trials. Ann Oncol 2025; 36:572-582. [PMID: 39986927 PMCID: PMC12034480 DOI: 10.1016/j.annonc.2025.01.017] [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: 11/16/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The SAKK 09/10 trial randomized biochemically recurrent prostate cancer patients to salvage radiation 64 Gy versus 70 Gy, and the NRG/RTOG 0126 randomized intermediate-risk prostate cancer patients to definitive radiation 70.2 Gy versus 79.2 Gy. We investigated a previously developed Post-Operative Radiation Therapy Outcomes Score (PORTOS) to identify preferential benefit from radiation dose escalation (DE). MATERIALS AND METHODS PORTOS was evaluated in patients enrolled in SAKK 09/10 and NRG/RTOG 0126 with available tissue that passed quality control (n = 226, 215). PORTOS was evaluated in the published post-operative groups in SAKK 09/10 and in tertiles in NRG/RTOG 0126 as cut-offs had not been established for biopsy samples and definitive radiation patients. Clinical and molecular correlates in a real-world dataset of 42 407 prostatectomy and 31 107 biopsy samples were also analyzed. RESULTS In SAKK 09/10, the biomarker-treatment interaction was statistically significant between PORTOS (lower versus higher) and treatment arm for clinical progression-free survival. Only patients in the higher PORTOS group benefited from DE. In NRG/RTOG 0126, in patients with a lower tertile PORTOS, there was no difference in Phoenix biochemical failure (BF). However, for patients in the average and higher tertile PORTOS range, there was a significant benefit for DE for Phoenix BF. An interaction test indicated a significant difference in benefit for DE between higher and lower PORTOS groups. PORTOS was not strongly associated with clinicopathological variables in either trial or the large real-world dataset. In the latter, PORTOS was modestly associated with hypoxia signatures and strongly associated with immune signatures and subtypes. CONCLUSION In the SAKK 09/10 and RTOG 0126 randomized controlled trials, we demonstrated that PORTOS can potentially identify a subset of patients who benefit from DE, a subgroup that cannot be identified using clinicopathological or prognostic variables. These results suggest that PORTOS could be used clinically as a predictor of radiation response.
Collapse
Affiliation(s)
- A Dal Pra
- Department of Radiation Oncology, University of Miami/Sylvester Comprehensive Cancer Center, Miami, USA.
| | - P Ghadjar
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - H M Ryu
- Veracyte Inc., San Diego, USA
| | | | - S Hayoz
- SAKK Coordinating Center, Bern, Switzerland
| | - J M Michalski
- Washington University School of Medicine, Saint Louis, USA
| | - D E Spratt
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, USA
| | - Y Liu
- Veracyte Inc., San Diego, USA
| | - C Schär
- SAKK Coordinating Center, Bern, Switzerland
| | - A M Berlin
- University Health Network-Princess Margaret Hospital, Toronto, Canada
| | - D R Zwahlen
- Department of Radiation Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - J P Simko
- University of California San Francisco, San Francisco, USA
| | - T Hölscher
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - J A Efstathiou
- Massachusetts General Hospital Cancer Center, Boston, USA
| | - B Polat
- Department of Radiation Oncology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - H M Sandler
- Cedars-Sinai Medical Center, Los Angeles, USA
| | - G Hildebrandt
- Department of Radiation Oncology, University Hospital Rostock, Rostock, Germany
| | | | - A-C Mueller
- Department of Radiation Oncology, Eberhard Karls University, Tübingen, Germany
| | - I S Dayes
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, Canada
| | - L Plasswilm
- Department of Radiation Oncology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | | | | - T G Karrison
- NRG Oncology Statistics and Data Management Center, Philadelphia, USA
| | | | - W A Hall
- Medical College of Wisconsin, Milwaukee, USA
| | - F Y Feng
- University of California San Francisco, San Francisco, USA
| | - A Pollack
- Department of Radiation Oncology, University of Miami/Sylvester Comprehensive Cancer Center, Miami, USA
| | - G N Thalmann
- Department of Urology, Bern University Hospital, Bern, Switzerland
| | - P L Nguyen
- Dana Farber Cancer Institute, Boston, USA
| | - D M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - P T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA.
| | - S G Zhao
- Department of Human Oncology, University of Wisconsin, Madison, USA.
| |
Collapse
|
2
|
Oseni SO, Naar C, Pavlović M, Asghar W, Hartmann JX, Fields GB, Esiobu N, Kumi-Diaka J. The Molecular Basis and Clinical Consequences of Chronic Inflammation in Prostatic Diseases: Prostatitis, Benign Prostatic Hyperplasia, and Prostate Cancer. Cancers (Basel) 2023; 15:3110. [PMID: 37370720 DOI: 10.3390/cancers15123110] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic inflammation is now recognized as one of the major risk factors and molecular hallmarks of chronic prostatitis, benign prostatic hyperplasia (BPH), and prostate tumorigenesis. However, the molecular mechanisms by which chronic inflammation signaling contributes to the pathogenesis of these prostate diseases are poorly understood. Previous efforts to therapeutically target the upstream (e.g., TLRs and IL1-Rs) and downstream (e.g., NF-κB subunits and cytokines) inflammatory signaling molecules in people with these conditions have been clinically ambiguous and unsatisfactory, hence fostering the recent paradigm shift towards unraveling and understanding the functional roles and clinical significance of the novel and relatively underexplored inflammatory molecules and pathways that could become potential therapeutic targets in managing prostatic diseases. In this review article, we exclusively discuss the causal and molecular drivers of prostatitis, BPH, and prostate tumorigenesis, as well as the potential impacts of microbiome dysbiosis and chronic inflammation in promoting prostate pathologies. We specifically focus on the importance of some of the underexplored druggable inflammatory molecules, by discussing how their aberrant signaling could promote prostate cancer (PCa) stemness, neuroendocrine differentiation, castration resistance, metabolic reprogramming, and immunosuppression. The potential contribution of the IL1R-TLR-IRAK-NF-κBs signaling molecules and NLR/inflammasomes in prostate pathologies, as well as the prospective benefits of selectively targeting the midstream molecules in the various inflammatory cascades, are also discussed. Though this review concentrates more on PCa, we envision that the information could be applied to other prostate diseases. In conclusion, we have underlined the molecular mechanisms and signaling pathways that may need to be targeted and/or further investigated to better understand the association between chronic inflammation and prostate diseases.
Collapse
Affiliation(s)
- Saheed Oluwasina Oseni
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Corey Naar
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Mirjana Pavlović
- Department of Computer and Electrical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Waseem Asghar
- Department of Computer and Electrical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - James X Hartmann
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, and I-HEALTH, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Nwadiuto Esiobu
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - James Kumi-Diaka
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
3
|
Zhong J, Frood R, McWilliam A, Davey A, Shortall J, Swinton M, Hulson O, West CM, Buckley D, Brown S, Choudhury A, Hoskin P, Henry A, Scarsbrook A. Prediction of prostate tumour hypoxia using pre-treatment MRI-derived radiomics: preliminary findings. LA RADIOLOGIA MEDICA 2023; 128:765-774. [PMID: 37198374 PMCID: PMC10264289 DOI: 10.1007/s11547-023-01644-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/26/2023] [Indexed: 05/19/2023]
Abstract
PURPOSE To develop a machine learning (ML) model based on radiomic features (RF) extracted from whole prostate gland magnetic resonance imaging (MRI) for prediction of tumour hypoxia pre-radiotherapy. MATERIAL AND METHODS Consecutive patients with high-grade prostate cancer and pre-treatment MRI treated with radiotherapy between 01/12/2007 and 1/08/2013 at two cancer centres were included. Cancers were dichotomised as normoxic or hypoxic using a biopsy-based 32-gene hypoxia signature (Ragnum signature). Prostate segmentation was performed on axial T2-weighted (T2w) sequences using RayStation (v9.1). Histogram standardisation was applied prior to RF extraction. PyRadiomics (v3.0.1) was used to extract RFs for analysis. The cohort was split 80:20 into training and test sets. Six different ML classifiers for distinguishing hypoxia were trained and tuned using five different feature selection models and fivefold cross-validation with 20 repeats. The model with the highest mean validation area under the curve (AUC) receiver operating characteristic (ROC) curve was tested on the unseen set, and AUCs were compared via DeLong test with 95% confidence interval (CI). RESULTS 195 patients were included with 97 (49.7%) having hypoxic tumours. The hypoxia prediction model with best performance was derived using ridge regression and had a test AUC of 0.69 (95% CI: 0.14). The test AUC for the clinical-only model was lower (0.57), but this was not statistically significant (p = 0.35). The five selected RFs included textural and wavelet-transformed features. CONCLUSION Whole prostate MRI-radiomics has the potential to non-invasively predict tumour hypoxia prior to radiotherapy which may be helpful for individualised treatment optimisation.
Collapse
Affiliation(s)
- Jim Zhong
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK.
| | - Russell Frood
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| | - Alan McWilliam
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Angela Davey
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Jane Shortall
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Martin Swinton
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Oliver Hulson
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| | - Catharine M West
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - David Buckley
- Biomedical Imaging, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sarah Brown
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research (LICTR), University of Leeds, Leeds, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Ann Henry
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Clinical Oncology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| | - Andrew Scarsbrook
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| |
Collapse
|
4
|
Cameron S, Deblois G, Hawley JR, Qamra A, Zhou S, Tonekaboni SAM, Murison A, Van Vliet R, Liu J, Locasale JW, Lupien M. Chronic hypoxia favours adoption to a castration-resistant cell state in prostate cancer. Oncogene 2023; 42:1693-1703. [PMID: 37020039 DOI: 10.1038/s41388-023-02680-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/10/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023]
Abstract
Predicting and treating recurrence in intermediate-risk prostate cancer patients remains a challenge despite having identified genomic instability [1] and hypoxia [2, 3] as risk factors. This underlies challenges in assigning the functional impact of these risk factors to mechanisms promoting prostate cancer progression. Here we show chronic hypoxia (CH), as observed in prostate tumours [4], leads to the adoption of an androgen-independent state in prostate cancer cells. Specifically, CH results in prostate cancer cells adopting transcriptional and metabolic alterations typical of castration-resistant prostate cancer cells. These changes include the increased expression of transmembrane transporters for the methionine cycle and related pathways leading to increased abundance of metabolites and expression of enzymes related to glycolysis. Targeting of the Glucose Transporter 1 (GLUT1) identified a dependency on glycolysis in androgen-independent cells. Overall, we identified a therapeutically targetable weakness in chronic hypoxia and androgen-independent prostate cancer. These findings may offer additional strategies for treatment development against hypoxic prostate cancer.
Collapse
Affiliation(s)
- Sarina Cameron
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada
| | - Genevieve Deblois
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada
- Institute for Research in Immunology and Cancer (IRIC), Faculty of Pharmacy, University of Montréal, Montréal, QC, H3T 1J4, Canada
| | - James R Hawley
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Aditi Qamra
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada
| | - Stanley Zhou
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Seyed Ali Madani Tonekaboni
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | - Romy Van Vliet
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada
| | - Juan Liu
- Duke University School of Medicine, Durham, NC, USA
| | | | - Mathieu Lupien
- Princess Margaret Cancer Research Centre, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Ontario Institute for Cancer Research, Toronto, ON, Canada.
| |
Collapse
|
5
|
A GdW10@PDA-CAT Sensitizer with High-Z Effect and Self-Supplied Oxygen for Hypoxic-Tumor Radiotherapy. Molecules 2021; 27:molecules27010128. [PMID: 35011360 PMCID: PMC8746738 DOI: 10.3390/molecules27010128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/13/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
Anticancer treatment is largely affected by the hypoxic tumor microenvironment (TME), which causes the resistance of the tumor to radiotherapy. Combining radiosensitizer compounds and O2 self-enriched moieties is an emerging strategy in hypoxic-tumor treatments. Herein, we engineered GdW10@PDA-CAT (K3Na4H2GdW10O36·2H2O, GdW10, polydopamine, PDA, catalase, CAT) composites as a radiosensitizer for the TME-manipulated enhancement of radiotherapy. In the composites, Gd (Z = 64) and W (Z = 74), as the high Z elements, make X-ray gather in tumor cells, thereby enhancing DNA damage induced by radiation. CAT can convert H2O2 to O2 and H2O to enhance the X-ray effect under hypoxic TME. CAT and PDA modification enhances the biocompatibility of the composites. Our results showed that GdW10@PDA-CAT composites increased the efficiency of radiotherapy in HT29 cells in culture. This polyoxometalates and O2 self-supplement composites provide a promising radiosensitizer for the radiotherapy field.
Collapse
|
6
|
Geng H, Ko HK, Pittsenbarger J, Harvey CT, Xue C, Liu Q, Wiens S, Kachhap SK, Beer TM, Qian DZ. HIF1 and ID1 Interplay Confers Adaptive Survival to HIF1α-Inhibition. Front Cell Dev Biol 2021; 9:724059. [PMID: 34820369 PMCID: PMC8606652 DOI: 10.3389/fcell.2021.724059] [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: 06/11/2021] [Accepted: 10/06/2021] [Indexed: 11/20/2022] Open
Abstract
Hypoxia is a universal pathological feature of solid tumors. Hypoxic tumor cells acquire metastatic and lethal phenotypes primarily through the activities of hypoxia-inducible factor 1 alpha (HIF1α). Therefore, HIF1α is considered as a promising therapeutic target. However, HIF inhibitors have not proven to be effective in clinical testing. The underlying mechanism is unclear. We report that oncogenic protein ID1 is upregulated in hypoxia by HIF1α shRNA or pharmacological inhibitors. In turn, ID1 supports tumor growth in hypoxia in vitro and in xenografts in vivo, conferring adaptive survival response and resistance. Mechanistically, ID1 proteins interfere HIF1-mediated gene transcription activation, thus ID1 protein degradation is accelerated by HIF1α-dependent mechanisms in hypoxia. Inhibitions of HIF1α rescues ID1, which compensates the loss of HIF1α by the upregulation of GLS2 and glutamine metabolism, thereby switching the metabolic dependency of HIF1α -inhibited cells from glucose to glutamine.
Collapse
Affiliation(s)
- Hao Geng
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Hyun-Kyung Ko
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Janet Pittsenbarger
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Christopher T Harvey
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Changhui Xue
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Qiong Liu
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Sadie Wiens
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Sushant K Kachhap
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Tomasz M Beer
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - David Z Qian
- Prostate Cancer Research Program, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
7
|
Ramotar M, Chua MLK, Truong H, Hosni A, Pintilie M, Davicioni E, Fleshner NE, Dicker AP, Bristow RG, He HH, van der Kwast T, Den RB, Berlin A. Subpathologies and genomic classifier for treatment individualization of post-prostatectomy radiotherapy. Urol Oncol 2021; 40:5.e1-5.e13. [PMID: 34538726 DOI: 10.1016/j.urolonc.2021.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/01/2021] [Accepted: 08/13/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE/OBJECTIVE Risk-stratification for post-prostatectomy radiotherapy (PORT) using conventional clinicopathologic indexes leads to substantial over- and under-treatment. Better patient selection could spare unnecessary toxicities and improve outcomes. We investigated the prognostic utility of unfavorable subpathologies intraductal carcinoma and cribriform architecture (IDC/CA), and a 22-gene Decipher genomic classifier (GC) in prostate cancer (PCa) patients receiving PORT. MATERIAL/METHODS A cohort of 302 men who received PORT at 2 academic institutions was pooled. PORT was predominately delivered as salvage (62% of cases); 20% received HT+PORT. Specimens were centrally reviewed for IDC/CA presence. In 104 cases, GC scores were determined. Endpoints were biochemical relapse-free (bRFR) and metastasis-free (mFR) rates. RESULTS After a median follow-up of 6.49-years, 135 (45%) and 40 (13%) men experienced biochemical relapse and metastasis, respectively. IDC/CA were identified in 160 (53%) of cases. Men harboring IDC/CA experienced inferior bRFR (HR 2.6, 95%CI 1.8-3.2, P<0.001) and mFR (HR 3.1, 95%CI 1.5-6.4, P = 0.0014). Patients with GC scores, 22 (21%) were stratified low-, 30 (29%) intermediate-, and 52 (50%) high-risk. GC low-risk was associated with superior bRFR (HR 0.25, 95%CI 0.1-0.5, P<0.001) and mFR (HR 0.15, 95%CI 0.03-0.8, P = 0.025). On multivariable analyses, IDC/CA and GC independently predicted for bRFR, corresponding to improved discrimination (C-index = 0.737 (95%CI 0.662-0.813)). CONCLUSIONS IDC/CA subpathologies and GC predict for biochemical relapse and metastasis beyond conventional clinicopathologic indexes in the PORT setting. Patients harboring IDC/CA are at higher risk of relapse after maximal local therapies, thus warranting consideration for treatment intensification strategies. Conversely, for men with absence of IDC/CA and low GC scores, de-intensification strategies could be explored.
Collapse
Affiliation(s)
- Matthew Ramotar
- Department of Radiation Oncology, University of Toronto; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Melvin L K Chua
- Department of Radiation Oncology, University of Toronto; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Divisions of Radiation Oncology and Medical Sciences, National Cancer Centre Singapore, Singapore; Oncology Academic Programme, Duke-NUS Medical School, Singapore
| | - Hong Truong
- Department of Urology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Ali Hosni
- Department of Radiation Oncology, University of Toronto; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Melania Pintilie
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Neil E Fleshner
- Division of Urology, University of Toronto; Mount Sinai Hospital; Princess Margaret Cancer Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Adam P Dicker
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Robert G Bristow
- Department of Radiation Oncology, University of Toronto; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Manchester Cancer Research Center, Manchester, United Kingdom
| | - Hansen H He
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Theo van der Kwast
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Robert B Den
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.
| | - Alejandro Berlin
- Department of Radiation Oncology, University of Toronto; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Techna Institute, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Lawrence MG, Porter LH, Clouston D, Murphy DG, Frydenberg M, Taylor RA, Risbridger GP. Knowing what's growing: Why ductal and intraductal prostate cancer matter. Sci Transl Med 2021; 12:12/533/eaaz0152. [PMID: 32132214 DOI: 10.1126/scitranslmed.aaz0152] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Prostate cancer is a common malignancy, but only some tumors are lethal. Accurately identifying these tumors will improve clinical practice and instruct research. Aggressive cancers often have distinctive pathologies, including intraductal carcinoma of the prostate (IDC-P) and ductal adenocarcinoma. Here, we review the importance of these pathologies because they are often overlooked, especially in genomics and preclinical testing. Pathology, genomics, and patient-derived models show that IDC-P and ductal adenocarcinoma accompany multiple markers of poor prognosis. Consequently, "knowing what is growing" will help translate preclinical research to pinpoint and treat high-risk prostate cancer in the clinic.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Laura H Porter
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | | | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3000, Australia.,Epworth HealthCare, Melbourne, VIC 3000, Australia
| | - Mark Frydenberg
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia.,Australian Urology Associates, Melbourne, VIC 3000, Australia.,Department of Urology, Cabrini Health, Malvern, VIC 3144, Australia
| | - Renea A Taylor
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC 3800, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia. .,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
9
|
Elming PB, Wittenborn TR, Busk M, Sørensen BS, Thomsen MBH, Strandgaard T, Dyrskjøt L, Nielsen S, Horsman MR. Refinement of an Established Procedure and Its Application for Identification of Hypoxia in Prostate Cancer Xenografts. Cancers (Basel) 2021; 13:2602. [PMID: 34073301 PMCID: PMC8198481 DOI: 10.3390/cancers13112602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND This pre-clinical study was designed to refine a dissection method for validating the use of a 15-gene hypoxia classifier, which was previously established for head and neck squamous cell carcinoma (HNSCC) patients, to identify hypoxia in prostate cancer. METHODS PC3 and DU-145 adenocarcinoma cells, in vitro, were gassed with various oxygen concentrations (0-21%) for 24 h, followed by real-time PCR. Xenografts were established in vivo, and the mice were injected with the hypoxic markers [18F]-FAZA and pimonidazole. Subsequently, tumors were excised, frozen, cryo-sectioned, and analyzed using autoradiography ([18F]-FAZA) and immunohistochemistry (pimonidazole); the autoradiograms used as templates for laser capture microdissection of hypoxic and non-hypoxic areas, which were lysed, and real-time PCR was performed. RESULTS In vitro, all 15 genes were increasingly up-regulated as oxygen concentrations decreased. With the xenografts, all 15 genes were up-regulated in the hypoxic compared to non-hypoxic areas for both cell lines, although this effect was greater in the DU-145. CONCLUSIONS We have developed a combined autoradiographic/laser-guided microdissection method with broad applicability. Using this approach on fresh frozen tumor material, thereby minimizing the degree of RNA degradation, we showed that the 15-gene hypoxia gene classifier developed in HNSCC may be applicable for adenocarcinomas such as prostate cancer.
Collapse
Affiliation(s)
- Pernille B. Elming
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Thomas R. Wittenborn
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Morten Busk
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Brita S. Sørensen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Mathilde Borg Houlberg Thomsen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Steffen Nielsen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Michael R. Horsman
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| |
Collapse
|
10
|
Abstract
Prostate cancer (PCa) is a clinically heterogeneous disease and has poor patient outcome when tumours progress to castration-resistant and metastatic states. Understanding the mechanistic basis for transition to late stage aggressive disease is vital for both assigning patient risk status in the localised setting and also identifying novel treatment strategies to prevent progression. Subregions of intratumoral hypoxia are found in all solid tumours and are associated with many biologic drivers of tumour progression. Crucially, more recent findings show the co-presence of hypoxia and genomic instability can confer a uniquely adverse prognosis in localised PCa patients. In-depth informatic and functional studies suggests a role for hypoxia in co-operating with oncogenic drivers (e.g. loss of PTEN) and suppressing DNA repair capacity to alter clonal evolution due to an aggressive mutator phenotype. More specifically, hypoxic suppression of homologous recombination represents a “contextual lethal“ vulnerability in hypoxic prostate tumours which could extend the application of existing DNA repair targeting agents such as poly-ADP ribose polymerase inhibitors. Further investigation is now required to assess this relationship on the background of existing genomic alterations relevant to PCa, and also characterise the role of hypoxia in driving early metastatic spread. On this basis, PCa patients with hypoxic tumours can be better stratified into risk categories and treated with appropriate therapies to prevent progression.
Collapse
Affiliation(s)
- Jack Ashton
- Translational Oncogenomics, CRUK Manchester Institute and CRUK Manchester Centre, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert Bristow
- Translational Oncogenomics, CRUK Manchester Institute and CRUK Manchester Centre, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
11
|
Bates M, Boland A, McDermott N, Marignol L. YB-1: The key to personalised prostate cancer management? Cancer Lett 2020; 490:66-75. [PMID: 32681926 DOI: 10.1016/j.canlet.2020.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
Y-box-binding protein 1 (YB-1) is a DNA/RNA binding protein increasingly implicated in the regulation of cancer cell biology. Normally located in the cytoplasm, nuclear localisation in prostate cancer is associated with more aggressive, potentially treatment-resistant disease. This is attributed to the ability of YB-1 to act as a transcription factor for various target genes associated with androgen receptor signalling, survival, DNA repair, proliferation, invasion, differentiation, angiogenesis and hypoxia. This review aims to examine the clinical potential of YB-1 in the detection and therapeutic management of prostate cancer.
Collapse
Affiliation(s)
- Mark Bates
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland
| | - Anna Boland
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland
| | - Niamh McDermott
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland
| | - Laure Marignol
- Translational Radiobiology and Molecular Oncology Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
12
|
Elevated HDAC activity and altered histone phospho-acetylation confer acquired radio-resistant phenotype to breast cancer cells. Clin Epigenetics 2020; 12:4. [PMID: 31900196 PMCID: PMC6942324 DOI: 10.1186/s13148-019-0800-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/23/2019] [Indexed: 12/18/2022] Open
Abstract
Background Poor-responsiveness of tumors to radiotherapy is a major clinical problem. Owing to the dynamic nature of the epigenome, the identification and targeting of potential epigenetic modifiers may be helpful to curb radio-resistance. This requires a detailed exploration of the epigenetic changes that occur during the acquirement of radio-resistance. Such an understanding can be applied for effective utilization of treatment adjuncts to enhance the efficacy of radiotherapy and reduce the incidence of tumor recurrence. Results This study explored the epigenetic alterations that occur during the acquirement of radio-resistance. Sequential irradiation of MCF7 breast cancer cell line up to 20 Gy generated a radio-resistant model. Micrococcal nuclease digestion demonstrated the presence of compact chromatin architecture coupled with decreased levels of histone PTMs H3K9ac, H3K27 ac, and H3S10pK14ac in the G0/G1 and mitotic cell cycle phases of the radio-resistant cells. Further investigation revealed that the radio-resistant population possessed high HDAC and low HAT activity, thus making them suitable candidates for HDAC inhibitor–based radio-sensitization. Treatment of radio-resistant cells with HDAC inhibitor valproic acid led to the retention of γH2AX and decreased H3S10p after irradiation. Additionally, an analysis of 38 human patient samples obtained from 8 different tumor types showed variable tumor HDAC activity, thus demonstrating inter-tumoral epigenetic heterogeneity in a patient population. Conclusion The study revealed that an imbalance of HAT and HDAC activities led to the loss of site-specific histone acetylation and chromatin compaction as breast cancer cells acquired radio-resistance. Due to variation in the tumor HDAC activity among patients, our report suggests performing a prior assessment of the tumor epigenome to maximize the benefit of HDAC inhibitor–based radio-sensitization. Graphical abstract ![]()
Collapse
|
13
|
Dünker N, Jendrossek V. Implementation of the Chick Chorioallantoic Membrane (CAM) Model in Radiation Biology and Experimental Radiation Oncology Research. Cancers (Basel) 2019; 11:cancers11101499. [PMID: 31591362 PMCID: PMC6826367 DOI: 10.3390/cancers11101499] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) is part of standard cancer treatment. Innovations in treatment planning and increased precision in dose delivery have significantly improved the therapeutic gain of radiotherapy but are reaching their limits due to biologic constraints. Thus, a better understanding of the complex local and systemic responses to RT and of the biological mechanisms causing treatment success or failure is required if we aim to define novel targets for biological therapy optimization. Moreover, optimal treatment schedules and prognostic biomarkers have to be defined for assigning patients to the best treatment option. The complexity of the tumor environment and of the radiation response requires extensive in vivo experiments for the validation of such treatments. So far in vivo investigations have mostly been performed in time- and cost-intensive murine models. Here we propose the implementation of the chick chorioallantoic membrane (CAM) model as a fast, cost-efficient model for semi high-throughput preclinical in vivo screening of the modulation of the radiation effects by molecularly targeted drugs. This review provides a comprehensive overview on the application spectrum, advantages and limitations of the CAM assay and summarizes current knowledge of its applicability for cancer research with special focus on research in radiation biology and experimental radiation oncology.
Collapse
Affiliation(s)
- Nicole Dünker
- Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| |
Collapse
|
14
|
Sun Y, Williams S, Byrne D, Keam S, Reynolds HM, Mitchell C, Wraith D, Murphy D, Haworth A. Association analysis between quantitative MRI features and hypoxia-related genetic profiles in prostate cancer: a pilot study. Br J Radiol 2019; 92:20190373. [PMID: 31356111 DOI: 10.1259/bjr.20190373] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To investigate the association between multiparametric MRI (mpMRI) imaging features and hypoxia-related genetic profiles in prostate cancer. METHODS In vivo mpMRI was acquired from six patients prior to radical prostatectomy. Sequences included T2 weighted (T2W) imaging, diffusion-weighted imaging, dynamic contrast enhanced MRI and blood oxygen-level dependent imaging. Imaging data were co-registered with histology using three-dimensional deformable registration methods. Texture features were extracted from T2W images and parametric maps from functional MRI. Full transcriptome genetic profiles were obtained using next generation sequencing from the prostate specimens. Pearson correlation coefficients were calculated between mpMRI data and hypoxia-related gene expression levels. Results were validated using glucose transporter one immunohistochemistry (IHC). RESULTS Correlation analysis identified 34 candidate imaging features (six from the mpMRI data and 28 from T2W texture features). The IHC validation showed that 16 out of the 28 T2W texture features achieved weak but significant correlations (p < 0.05). CONCLUSIONS Weak associations between mpMRI features and hypoxia gene expressions were found. This indicates the potential use of MRI in assessing hypoxia status in prostate cancer. Further validation is required due to the low correlation levels. ADVANCES IN KNOWLEDGE This is a pilot study using radiogenomics approaches to address hypoxia within the prostate, which provides an opportunity for hypoxia-guided selective treatment techniques.
Collapse
Affiliation(s)
- Yu Sun
- The University of Sydney, Sydney, New South Wales, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Scott Williams
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - David Byrne
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Simon Keam
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Hayley M Reynolds
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Darren Wraith
- Queensland University of Technology, Brisbane, Queensland, Australia
| | - Declan Murphy
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Annette Haworth
- The University of Sydney, Sydney, New South Wales, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Marignol L. Predicting Treatment Outcomes: The Case for Hypoxia Gene Signatures. EBioMedicine 2019; 32:3-4. [PMID: 29776747 PMCID: PMC6020707 DOI: 10.1016/j.ebiom.2018.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 12/01/2022] Open
Affiliation(s)
- Laure Marignol
- Translational Radiobiology and Oncology, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy and Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
16
|
Personalising Prostate Radiotherapy in the Era of Precision Medicine: A Review. J Med Imaging Radiat Sci 2018; 49:376-382. [PMID: 30514554 DOI: 10.1016/j.jmir.2018.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/27/2017] [Accepted: 01/18/2018] [Indexed: 12/14/2022]
Abstract
Prostate cancer continues to be the most commonly diagnosed cancer among Canadian men. The introduction of routine screening and advanced treatment options have allowed for a decrease in prostate cancer-related mortality, but outcomes following treatment continue to vary widely. In addition, the overtreatment of indolent prostate cancers causes unnecessary treatment toxicities and burdens health care systems. Accurate identification of patients who should undergo aggressive treatment, and those which should be managed more conservatively, needs to be implemented. More tumour and patient information is needed to stratify patients into low-, intermediate-, and high-risk groups to guide treatment options. This paper reviews the current literature on personalised prostate cancer management, including targeting tumour hypoxia, genomic and radiomic prognosticators, and radiobiological tumour targeting. A review of the current applications and future directions for the use of big data in radiation therapy is also presented. Prostate cancer management has a lot to gain from the implementation of personalised medicine into practice. Using specific tumour and patient characteristics to personalise prostate radiotherapy in the era of precision medicine will improve survival, decrease unnecessary toxicities, and minimise the heterogeneity of outcomes following treatment.
Collapse
|
17
|
Atorvastatin enhances radiosensitivity in hypoxia-induced prostate cancer cells related with HIF-1α inhibition. Biosci Rep 2017; 37:BSR20170340. [PMID: 28760843 PMCID: PMC5563539 DOI: 10.1042/bsr20170340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/10/2017] [Accepted: 07/31/2017] [Indexed: 12/26/2022] Open
Abstract
Hypoxia could enhance radioresistance in prostate cancer cells through up-regulating HIF-1α, which could be inhibited by statins in several cancer cells. However, this effect of statins in prostate cancer remains unclear. In the present study, we aim to investigate the effect of atorvastatin on HIF-1α expression and radiosensitivity in prostate cancer cells. The hypoxia-induced human prostate cancer PC3 cells were generated by incubating with 5% O2 for 24 h. The cell viability and apoptosis were respectively analyzed by cell counting kit-8 (CCK-8) assay and flow cytometry. The HIF-1α protein expression was assessed by Western blotting. HIF-1α expression in PC3 cells was significantly increased after incubating with 5% O2 for 24 h. The viability of hypoxia-induced PC3 cells was inhibited by a higher dose of irradiation than control cells. The viability of hypoxia-induced PC3 cells were inhibited by astorvastatin with a higher concentration than control cells. Astorvastatin reduced the HIF-1α protein expression in hypoxia-induced PC3 cells, and induced apoptosis of both control and hypoxia-induced cells with and without irradiation. Atorvastatin could enhance radiosensitivity in hypoxia-induced prostate cancer cells, which may be related with inhibition of HIF-1α protein.
Collapse
|
18
|
Berlin A, Castro-Mesta JF, Rodriguez-Romo L, Hernandez-Barajas D, González-Guerrero JF, Rodríguez-Fernández IA, González-Conchas G, Verdines-Perez A, Vera-Badillo FE. Prognostic role of Ki-67 score in localized prostate cancer: A systematic review and meta-analysis. Urol Oncol 2017. [PMID: 28648414 DOI: 10.1016/j.urolonc.2017.05.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Ki-67 for quantifying tumor proliferation is widely used. In localized prostate cancer (PCa), despite a suggested predictive role of Ki-67 for outcomes after therapies, it has not been incorporated into clinical practice. Herein, we conduct a systematic review and meta-analysis of the literature reporting the association of Ki-67 and disease outcomes in PCa treated radically. METHODS Medline and EMBASE databases were searched without date or language restrictions, using "KI67" and "prostate cancer" MeSH terms. Studies reporting Ki-67 association with clinical outcomes (disease-free survival [DFS], biochemical failure-free survival, rate of distant metastases [DM], disease-specific survival [DSS], or overall survival [OS], or all of these) in patients with PCa managed actively were included, and relevant data extracted by 2 independent reviewers. Odds ratios (OR) were weighted and pooled in a meta-analysis using Mantel-Haenszel random-effect modeling. RESULTS Twenty-one studies comprising 5,419 patients met eligibility for analysis, and 67.6% of patients had low Ki-67. Mean Ki-67 was 6.14%. High Ki-67 was strongly associated with worse clinical outcomes. DFS was better in those patients with low Ki-67 at 5 and 10 years (OR = 0.32, 95% CI: 0.23-0.44, P<0.00001; OR = 0.31, 95% CI: 0.20-0.48, P<0.00001). Similarly, low Ki-67 was related to improved DSS at 5 and 10 years (OR = 0.15, 95% CI: 0.10-0.21, P<0.00001; OR = 0.16, 95% CI: 0.06-0.40, P<0.00001). Association between low Ki-67 scores with improved OS (OR = 0.47; 95% CI: 0.37-0.61; P<0.00001) and high Ki-67 scores with DM at 5 years (OR = 4.07; 95% CI: 2.52-6.58; P<0.00001) was consistently observed. CONCLUSIONS High Ki-67 expression in localized PCa is a factor of poor prognosis for DSS, biochemical failure-free survival, DFS, DM, and OS after curative-intent treatments. Incorporation into clinical routine of this widely available and standardized biomarker should be strongly considered.
Collapse
Affiliation(s)
- Alejandro Berlin
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Julio F Castro-Mesta
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Laura Rodriguez-Romo
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - David Hernandez-Barajas
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Juan F González-Guerrero
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Iván A Rodríguez-Fernández
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Galileo González-Conchas
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Adrian Verdines-Perez
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Francisco E Vera-Badillo
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México; Department of Medical Oncology, Faculty of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
19
|
Challapalli A, Carroll L, Aboagye EO. Molecular mechanisms of hypoxia in cancer. Clin Transl Imaging 2017; 5:225-253. [PMID: 28596947 PMCID: PMC5437135 DOI: 10.1007/s40336-017-0231-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/21/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Hypoxia is a condition of insufficient oxygen to support metabolism which occurs when the vascular supply is interrupted, or when a tumour outgrows its vascular supply. It is a negative prognostic factor due to its association with an aggressive tumour phenotype and therapeutic resistance. This review provides an overview of hypoxia imaging with Positron emission tomography (PET), with an emphasis on the biological relevance, mechanism of action, highlighting advantages, and limitations of the currently available hypoxia radiotracers. METHODS A comprehensive PubMed literature search was performed, identifying articles relating to biological significance and measurement of hypoxia, MRI methods, and PET imaging of hypoxia in preclinical and clinical settings, up to December 2016. RESULTS A variety of approaches have been explored over the years for detecting and monitoring changes in tumour hypoxia, including regional measurements with oxygen electrodes placed under CT guidance, MRI methods that measure either oxygenation or lactate production consequent to hypoxia, different nuclear medicine approaches that utilise imaging agents the accumulation of which is inversely related to oxygen tension, and optical methods. The advantages and disadvantages of these approaches are reviewed, along with individual strategies for validating different imaging methods. PET is the preferred method for imaging tumour hypoxia due to its high specificity and sensitivity to probe physiological processes in vivo, as well as the ability to provide information about intracellular oxygenation levels. CONCLUSION Even though hypoxia could have significant prognostic and predictive value in the clinic, the best method for hypoxia assessment has in our opinion not been realised.
Collapse
Affiliation(s)
- Amarnath Challapalli
- Department of Clinical Oncology, Bristol Cancer Institute, Horfield Road, Bristol, United Kingdom
| | - Laurence Carroll
- Department of Surgery and Cancer, Imperial College, GN1, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W120NN United Kingdom
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College, GN1, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W120NN United Kingdom
| |
Collapse
|
20
|
Raziee H, Moraes FY, Murgic J, Chua MLK, Pintilie M, Chung P, Ménard C, Bayley A, Gospodarowicz M, Warde P, Craig T, Catton C, Bristow RG, Jaffray DA, Berlin A. Improved outcomes with dose escalation in localized prostate cancer treated with precision image-guided radiotherapy. Radiother Oncol 2017; 123:459-465. [PMID: 28434799 DOI: 10.1016/j.radonc.2017.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/12/2017] [Accepted: 04/01/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND PURPOSE Dose-escalated radiotherapy (DE) improves outcomes in localized prostate cancer (PCa). The impact of DE in the context of image-guided radiotherapy (IGRT) remains unknown. Herein, we determined outcomes of three sequential cohorts treated with progressive DE-IGRT. MATERIALS AND METHODS We analyzed data from 1998 to 2012. Patients treated with radical radiotherapy were included, with three sequential institutional schedules: (A) 75.6Gy, (B) 79.8Gy, (C) 78Gy, with 1.8, 1.9 and 2Gy/fraction, respectively. IGRT consisted of fiducial markers and daily EPID (A, B) or CBCT (C). RESULTS 961 patients were included, with median follow-up of 6.1y. 30.5%, 32.6% and 36.9% were treated in A, B and C, respectively. Risk category distribution was 179 (18.6%) low-, 653 (67.9%) intermediate- and 129 (13.5%) high-risk. PSA, T-category, androgen deprivation use and risk distribution were similar among groups. BCR (biochemical recurrence) was different (p<0.001) between A, B and C with 5-year rates of 23%, 17% and 9%, respectively (HR 2.68 [95% CI 1.87-3.85] and 1.92 [95% CI 1.33-2.78] for A and B compared to C, respectively). Findings were most significant in the intermediate-risk category. Metastasis, cause-specific-death and toxicities were not different between cohorts. CONCLUSION Our findings suggest continuous BCR improvement with progressive DE-IGRT. Prospective validation considering further DE with IGRT seems warranted.
Collapse
Affiliation(s)
- Hamid Raziee
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Fabio Y Moraes
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Jure Murgic
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Melvin L K Chua
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Melania Pintilie
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Peter Chung
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Cynthia Ménard
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada; Département de radio-oncologie, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
| | - Andrew Bayley
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Mary Gospodarowicz
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Padraig Warde
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Tim Craig
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Charles Catton
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Robert G Bristow
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - David A Jaffray
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada
| | - Alejandro Berlin
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Department of Radiation Oncology, University of Toronto, Canada.
| |
Collapse
|
21
|
Stapleton S, Jaffray D, Milosevic M. Radiation effects on the tumor microenvironment: Implications for nanomedicine delivery. Adv Drug Deliv Rev 2017; 109:119-130. [PMID: 27262923 DOI: 10.1016/j.addr.2016.05.021] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/22/2016] [Accepted: 05/24/2016] [Indexed: 01/24/2023]
Abstract
The tumor microenvironment has an important influence on cancer biological and clinical behavior and radiation treatment (RT) response. However, RT also influences the tumor microenvironment in a complex and dynamic manner that can either reinforce or inhibit this response and the likelihood of long-term disease control in patients. It is increasingly evident that the interplay between RT and the tumor microenvironment can be exploited to enhance the accumulation and intra-tumoral distribution of nanoparticles, mediated by changes to the vasculature and stroma with secondary effects on hypoxia, interstitial fluid pressure (IFP), solid tissue pressure (STP), and the recruitment and activation of bone marrow-derived myeloid cells (BMDCs). The use of RT to modulate nanoparticle drug delivery offers an exciting opportunity to improve antitumor efficacy. This review explores the interplay between RT and the tumor microenvironment, and the integrated effects on nanoparticle drug delivery and efficacy.
Collapse
Affiliation(s)
- Shawn Stapleton
- Radiation Medicine Program, Princess Margaret Cancer Centre and University Health Network, Toronto, ON, Canada
| | - David Jaffray
- Radiation Medicine Program, Princess Margaret Cancer Centre and University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Michael Milosevic
- Radiation Medicine Program, Princess Margaret Cancer Centre and University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
22
|
Biomarkers of Outcome in Patients With Localized Prostate Cancer Treated With Radiotherapy. Semin Radiat Oncol 2017; 27:11-20. [DOI: 10.1016/j.semradonc.2016.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
23
|
Abstract
Tumours contain multiple different cell populations, including cells derived from the bone marrow as well as cancer-associated fibroblasts and various stromal populations including the vasculature. The microenvironment of the tumour cells plays a significant role in the response of the tumour to radiation treatment. Low levels of oxygen (hypoxia) caused by the poorly organized vasculature in tumours have long been known to affect radiation response; however, other aspects of the microenvironment may also play important roles. This article reviews some of the old literature concerning tumour response to irradiation and relates this to current concepts about the role of the tumour microenvironment in tumour response to radiation treatment. Included in the discussion are the role of cancer stem cells, radiation damage to the vasculature and the potential for radiation to enhance immune activity against tumour cells. Radiation treatment can cause a significant influx of bone marrow-derived cell populations into both normal tissues and tumours. Potential roles of such cells may include enhancing vascular recovery as well as modulating immune reactivity.
Collapse
Affiliation(s)
- Richard P Hill
- 1 Ontario Cancer Institute, Princess Margaret Cancer Centre, Toronto, ON, Canada.,2 Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Memon D, Dawson K, Smowton CSF, Xing W, Dive C, Miller CJ. Hypoxia-driven splicing into noncoding isoforms regulates the DNA damage response. NPJ Genom Med 2016; 1:16020. [PMID: 28480052 PMCID: PMC5417364 DOI: 10.1038/npjgenmed.2016.20] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 12/26/2022] Open
Abstract
Tumour hypoxia is associated with poor patient outcome and resistance to therapy. It is accompanied by widespread changes in gene expression mediated largely through the transcription factors HIF1/2/3α. Hypoxia impacts on multiple pathways throughout the cell and has widespread effects on phenotype. Here we use sample-specific annotation approaches to determine the changes in transcript architecture that arise as result of alternative splicing in hypoxic cells. Using in vivo data generated from a time course in reduced oxygenation we identified genome-wide switching between coding and noncoding isoforms, including a significant number of components of the DNA damage response pathway. Notably, HDAC6, a master regulator of the cytotoxic response, and TP53BP1, which sits at the nexus of the double-strand break repair pathway, both underwent a marked transition towards an intron-retention pattern with a concomitant decline in protein levels. These transitions from coding to noncoding isoforms were recapitulated in a large and independent cohort of 499 colorectal samples taken from The Cancer Genome Atlas (TCGA). The set of altered genes was enriched for multiple components of the Fanconi Anaemia, nucleotide excision and double-strand break repair pathways, and together correlating with tumour status at last contact. Altogether, these data demonstrate a new role for hypoxia-driven alternative splicing in regulating DNA damage response, and highlight the importance of considering alternative splicing as a critical factor in our understanding of human disease.
Collapse
Affiliation(s)
- Danish Memon
- RNA Biology Group, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Keren Dawson
- RNA Biology Group, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Christopher SF Smowton
- Scientific Computing Team, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Wei Xing
- Scientific Computing Team, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Crispin J Miller
- RNA Biology Group, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| |
Collapse
|
25
|
Sun Y, Jia X, Hou L, Liu X. Screening of Differently Expressed miRNA and mRNA in Prostate Cancer by Integrated Analysis of Transcription Data. Urology 2016; 94:313.e1-6. [PMID: 27179774 DOI: 10.1016/j.urology.2016.04.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 03/01/2016] [Accepted: 04/28/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The purpose of this study was to screen aberrantly expressed miRNAs and genes in prostate cancer (PCA), and further uncover the underlying mechanisms for the development of PCA. MATERIALS AND METHODS We searched the Gene Expression Omnibus database for miRNA and gene expression datasets of PCA, and then separately integrated miRNA and gene expression datasets to identify miRNA and gene expression profiles in PCA. Target genes of differentially expressed miRNAs were predicted through miRWalk database. We matched these target genes with the list of differentially expressed genes to identify miRNA-target gene pairs whose expression was inversely correlated. The function of these target genes was annotated. RESULTS Twenty-nine differentially expressed miRNAs and 946 differentially expressed genes were identified between PCA and normal control. Seven hundred fifty-one miRNA-target gene pairs that showed inverse expression in PCA were obtained to establish a regulatory network. In this regulatory network, 10 genes (BCL2, BNC2, CCND2, EPM2A, MRAS, NAV2, RASL12, STK33, TCEAL1, WWC2) were co-regulated by 5 miRNAs (hsa-miR-106b, hsa-miR-130b, hsa-miR-93, hsa-miR-153, hsa-miR-182). The expression of hsa-miR-182 was significantly associated with PCA survival through the online validation tool of SurvMicro, suggesting the potential use as a diagnostic or prognostic biomarker in PCA. CONCLUSION This integrated analysis was performed to infer new miRNA regulation activities, which provides insights into the understanding of underlying molecular mechanisms of PCA, and guides for exploration of novel therapeutic targets.
Collapse
Affiliation(s)
- Yanan Sun
- Department of Obstetrics and Gynecology, Bethune International Peace Hospital of PLA, Shijiazhuang, Hebei, China
| | - Xiaopeng Jia
- Department of Urology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Lianguo Hou
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xing Liu
- Orthopaedic Trauma Department 2, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| |
Collapse
|
26
|
Raziee H, Berlin A. Gaps between Evidence and Practice in Postoperative Radiotherapy for Prostate Cancer: Focus on Toxicities and the Effects on Health-Related Quality of Life. Front Oncol 2016; 6:70. [PMID: 27047800 PMCID: PMC4805642 DOI: 10.3389/fonc.2016.00070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/10/2016] [Indexed: 12/24/2022] Open
Abstract
Adjuvant radiotherapy (ART) after prostatectomy for patients with high-risk features [extracapsular extension (ECE), seminal vesicle invasion (SVI), and positive margin] has been shown to be associated with improved biochemical disease-free survival in three large randomized trials and with improved overall survival in one. Similarly, salvage radiotherapy (SRT) can effectively achieve biochemical control in a significant proportion of patients with a rising PSA after surgery. Nonetheless, both approaches of postoperative RT remain highly underutilized. This might be partly due to concerns with overtreatment inherent to adjuvant approaches, and/or hesitance about causing radiation toxicities and their subsequent effects on the patient's quality of life. Herein, we review the literature lending evidence to these arguments. We show recent series of ART/SRT and their low rates of acute and long-term toxicities, translating only in transient decline in quality-of-life (QoL) outcomes. We conclude that concerns with side effects should not preclude the recommendation of an effective and curative-intent therapy for men with prostate cancer initially treated with radical surgery.
Collapse
Affiliation(s)
- Hamid Raziee
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Alejandro Berlin
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
27
|
Prostate cancer radiation therapy: A physician’s perspective. Phys Med 2016; 32:438-45. [DOI: 10.1016/j.ejmp.2016.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/27/2016] [Accepted: 02/17/2016] [Indexed: 02/07/2023] Open
|
28
|
Eke I, Makinde AY, Aryankalayil MJ, Ahmed MM, Coleman CN. Comprehensive molecular tumor profiling in radiation oncology: How it could be used for precision medicine. Cancer Lett 2016; 382:118-126. [PMID: 26828133 DOI: 10.1016/j.canlet.2016.01.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/16/2022]
Abstract
New technologies enabling the analysis of various molecules, including DNA, RNA, proteins and small metabolites, can aid in understanding the complex molecular processes in cancer cells. In particular, for the use of novel targeted therapeutics, elucidation of the mechanisms leading to cell death or survival is crucial to eliminate tumor resistance and optimize therapeutic efficacy. While some techniques, such as genomic analysis for identifying specific gene mutations or epigenetic testing of promoter methylation, are already in clinical use, other "omics-based" assays are still evolving. Here, we provide an overview of the current status of molecular profiling methods, including promising research strategies, as well as possible challenges, and their emerging role in radiation oncology.
Collapse
Affiliation(s)
- Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Adeola Y Makinde
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mansoor M Ahmed
- Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| |
Collapse
|
29
|
Dhani N, Fyles A, Hedley D, Milosevic M. The clinical significance of hypoxia in human cancers. Semin Nucl Med 2015; 45:110-21. [PMID: 25704384 DOI: 10.1053/j.semnuclmed.2014.11.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hypoxia is present to some extent in most solid malignant human cancers because of an imbalance between the limited oxygen delivery capacity of the abnormal vasculature and the high oxygen consumption of tumor cells. This drives a complex and dynamic compensatory response to enable continued cell survival, including genomic changes leading to selection of hypoxia-adapted cells with a propensity to invade locally, metastasize, and recur following surgery or radiotherapy. There is indisputable clinical evidence from numerous observational and therapeutic studies across a range of tumor types to implicate hypoxia as a key determinant of cancer behavior and treatment outcome. Despite this, hypoxia-targeted treatment has failed to influence clinical practice. This is explained, in part, by emerging findings to indicate that hypoxia is not equally important in all patients even when present to the same extent. The impact of hypoxia on patient outcome and the benefit of hypoxia-targeted treatments are greatest in situations where hypoxia is a primary biological driver of disease behavior-patients with tumors having a "hypoxic driver" phenotype. The challenge for the clinical and scientific communities moving forward is to develop robust precision cancer medicine strategies for identifying these patients in the setting of other etiologic, genomic, and host-tumor factors, considering not only the state of the tumor at diagnosis but also changing patient and tumor characteristics over time.
Collapse
Affiliation(s)
- Neesha Dhani
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Anthony Fyles
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - David Hedley
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Michael Milosevic
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada.
| |
Collapse
|
30
|
Wilkins A, Dearnaley D, Somaiah N. Genomic and Histopathological Tissue Biomarkers That Predict Radiotherapy Response in Localised Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:238757. [PMID: 26504789 PMCID: PMC4609338 DOI: 10.1155/2015/238757] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/24/2015] [Indexed: 12/16/2022]
Abstract
Localised prostate cancer, in particular, intermediate risk disease, has varied survival outcomes that cannot be predicted accurately using current clinical risk factors. External beam radiotherapy (EBRT) is one of the standard curative treatment options for localised disease and its efficacy is related to wide ranging aspects of tumour biology. Histopathological techniques including immunohistochemistry and a variety of genomic assays have been used to identify biomarkers of tumour proliferation, cell cycle checkpoints, hypoxia, DNA repair, apoptosis, and androgen synthesis, which predict response to radiotherapy. Global measures of genomic instability also show exciting capacity to predict survival outcomes following EBRT. There is also an urgent clinical need for biomarkers to predict the radiotherapy fraction sensitivity of different prostate tumours and preclinical studies point to possible candidates. Finally, the increased resolution of next generation sequencing (NGS) is likely to enable yet more precise molecular predictions of radiotherapy response and fraction sensitivity.
Collapse
Affiliation(s)
- Anna Wilkins
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - David Dearnaley
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Navita Somaiah
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
- Division of Cancer Biology, The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
31
|
|
32
|
Sørensen BS, Knudsen A, Wittrup CF, Nielsen S, Aggerholm-Pedersen N, Busk M, Horsman M, Høyer M, Bouchelouche PN, Overgaard J, Alsner J. The usability of a 15-gene hypoxia classifier as a universal hypoxia profile in various cancer cell types. Radiother Oncol 2015; 116:346-51. [DOI: 10.1016/j.radonc.2015.06.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/19/2015] [Accepted: 06/30/2015] [Indexed: 02/05/2023]
|
33
|
Berlin A, Lalonde E, Sykes J, Zafarana G, Chu KC, Ramnarine VR, Ishkanian A, Sendorek DHS, Pasic I, Lam WL, Jurisica I, van der Kwast T, Milosevic M, Boutros PC, Bristow RG. NBN gain is predictive for adverse outcome following image-guided radiotherapy for localized prostate cancer. Oncotarget 2015; 5:11081-90. [PMID: 25415046 PMCID: PMC4294365 DOI: 10.18632/oncotarget.2404] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 08/26/2014] [Indexed: 12/31/2022] Open
Abstract
Despite the use of clinical prognostic factors (PSA, T-category and Gleason score), 20-60% of localized prostate cancers (PCa) fail primary local treatment. Herein, we determined the prognostic importance of main sensors of the DNA damage response (DDR): MRE11A, RAD50, NBN, ATM, ATR and PRKDC. We studied copy number alterations in DDR genes in localized PCa treated with image-guided radiotherapy (IGRT; n=139) versus radical prostatectomy (RadP; n=154). In both cohorts, NBN gains were the most frequent genomic alteration (14.4 and 11% of cases, respectively), and were associated with overall tumour genomic instability (p<0.0001). NBN gains were the only significant predictor of 5yrs biochemical relapse-free rate (bRFR) following IGRT (46% versus 77%; p=0.00067). On multivariate analysis, NBN gain remained a significant independent predictor of bRFR after adjusting for known clinical prognostic variables (HR=3.28, 95% CI 1.56–6.89, Wald p-value=0.0017). No DDR-sensing gene was prognostic in the RadP cohort. In vitro studies correlated NBN gene overexpression with PCa cells radioresistance. In conclusion, NBN gain predicts for decreased bRFR in IGRT, but not in RadP patients. If validated independently, Nibrin gains may be the first PCa predictive biomarker to facilitate local treatment decisions using precision medicine approaches with surgery or radiotherapy.
Collapse
Affiliation(s)
- Alejandro Berlin
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada. Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Emilie Lalonde
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada. Informatics and Bio-Computing, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Jenna Sykes
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada
| | - Gaetano Zafarana
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada
| | - Kenneth C Chu
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada. Informatics and Bio-Computing, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Varune R Ramnarine
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Adrian Ishkanian
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada. Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Dorota H S Sendorek
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada
| | - Ivan Pasic
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada
| | - Wan L Lam
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada. Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Igor Jurisica
- The Techna Institute, University Health Network, Toronto, ON, Canada
| | - Theo van der Kwast
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada
| | - Michael Milosevic
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada. Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Paul C Boutros
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada. Informatics and Bio-Computing, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Robert G Bristow
- Departments of Radiation Oncology, Medical Biophysics, Medical Oncology, Laboratory Medicine and Pathology, Pharmacology & Toxicology and Biostatistics, Computer Science, University of Toronto, Toronto, ON, Canada. Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
34
|
|
35
|
Locke JA, Dal Pra A, Supiot S, Warde P, Bristow RG. Synergistic action of image-guided radiotherapy and androgen deprivation therapy. Nat Rev Urol 2015; 12:193-204. [PMID: 25800395 DOI: 10.1038/nrurol.2015.50] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The combined use of androgen deprivation therapy (ADT) and image-guided radiotherapy (IGRT) can improve overall survival in aggressive, localized prostate cancer. However, owing to the adverse effects of prolonged ADT, it is imperative to identify the patients who would benefit from this combined-modality therapy relative to the use of IGRT alone. Opportunities exist for more personalized approaches in treating aggressive, locally advanced prostate cancer. Biomarkers--such as disseminated tumour cells, circulating tumour cells, genomic signatures and molecular imaging techniques--could identify the patients who are at greatest risk for systemic metastases and who would benefit from the addition of systemic ADT. By contrast, when biomarkers of systemic disease are not present, treatment could proceed using local IGRT alone. The choice of drug, treatment duration and timing of ADT relative to IGRT could be predicated on these personalized approaches to prostate cancer medicine. These novel treatment intensification and reduction strategies could result in improved prostate-cancer-specific survival and overall survival, without incurring the added expense of metabolic syndrome and other adverse effects of ADT in all patients.
Collapse
Affiliation(s)
- Jennifer A Locke
- Department of Urologic Sciences, University of British Columbia, Gordon &Leslie Diamond Health Care Centre, Level 6, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada
| | - Alan Dal Pra
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 4, CH-3010 Bern, Switzerland
| | - Stéphane Supiot
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes-St-Herblain, 8 quai Moncousu, BP 70721, 44000 Nantes, France
| | - Padraig Warde
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - Robert G Bristow
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
36
|
Korpela E, Vesprini D, Liu SK. MicroRNA in radiotherapy: miRage or miRador? Br J Cancer 2015; 112:777-82. [PMID: 25611301 PMCID: PMC4453960 DOI: 10.1038/bjc.2015.6] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 02/07/2023] Open
Abstract
At least half of all cancer patients will receive radiation therapy. Tumour radioresistance, or the failure to control certain tumours with this treatment, can result in locoregional recurrence; thus there is great interest in understanding the underlying biology and developing strategies to overcome this problem. The expanding investigation of microRNA in cancer suggests that these regulatory factors can influence the DNA damage response, the microenvironment and survival pathways, among other processes, and thereby may affect tumour radioresistance. As microRNA are readily detectable in tumours and biofluids, they hold promise as predictive biomarkers for therapy response and prognosis. This review highlights the current insights on the major ways that microRNA may contribute to tumour radiation response and whether their levels reflect treatment success. We conclude by applying the potential framework of future roles of miR in personalised radiotherapy using prostate cancer clinical management as an example.
Collapse
Affiliation(s)
- E Korpela
- Biological Sciences, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 1L7
| | - D Vesprini
- Biological Sciences, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada M5T 1P5
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5
| | - S K Liu
- Biological Sciences, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 1L7
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada M5T 1P5
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5
| |
Collapse
|
37
|
Berlin A, Dal Pra A, Bristow RG. Pre-radiotherapy identification of individual genomic profile to avoid, by resort to customized radiosensitizers, the risk of radioresistance development in patients with localized prostate cancer: author reply. Br J Radiol 2015; 88:20140701. [DOI: 10.1259/bjr.20140701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
38
|
Alberti C. Pre-radiotherapy identification of individual genomic profile to avoid, by resort to customized radiosensitizers, the risk of radioresistance development in patients with localized prostate cancer. Br J Radiol 2014; 88:20140630. [PMID: 25363844 DOI: 10.1259/bjr.20140630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
39
|
[What is the level of evidence of new techniques in prostate cancer radiotherapy?]. Cancer Radiother 2014; 18:501-8. [PMID: 25192625 DOI: 10.1016/j.canrad.2014.06.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 06/23/2014] [Accepted: 06/29/2014] [Indexed: 12/30/2022]
Abstract
Prostate cancer radiotherapy has evolved from the old 2D technique to conformal, and then to intensity-modulated radiation therapy (IMRT) and stereotactic radiotherapy. At the same time, image-guidance (IGRT) is routinely used. New techniques such as protontherapy or carbontherapy are being developed with the objective of increased efficacy, decreased treatment duration, toxicity or cost. This review summarizes the evidence-based medicine of new technologies in the treatment of prostate cancer.
Collapse
|
40
|
Affiliation(s)
- K M Prise
- Centre for Cancer Research and Cell Biology, Queens University Belfast, Belfast, UK
| | | |
Collapse
|