1
|
Yao K, Zhang R, Li L, Liu M, Feng S, Yan H, Zhang Z, Xie D. The signature of cuproptosis-related immune genes predicts the tumor microenvironment and prognosis of prostate adenocarcinoma. Front Immunol 2023; 14:1181370. [PMID: 37600770 PMCID: PMC10433769 DOI: 10.3389/fimmu.2023.1181370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background Cuproptosis plays a crucial role in cancer, and different subtypes of cuproptosis have different immune profiles in prostate adenocarcinoma (PRAD). This study aimed to investigate immune genes associated with cuproptosis and develop a risk model to predict prognostic characteristics and chemotherapy/immunotherapy responses of patients with PRAD. Methods The CIBERSORT algorithm was used to evaluate the immune and stromal scores of patients with PRAD in The Cancer Genome Atlas (TCGA) cohort. Validation of differentially expressed genes DLAT and DLD in benign and malignant tissues by immunohistochemistry, and the immune-related genes of DLAT and DLD were further screened. Univariable Cox regression were performed to select key genes. Least absolute shrinkage and selection operator (LASSO)-Cox regression analyse was used to develop a risk model based on the selected genes. The model was validated in the TCGA, Memorial Sloan-Kettering Cancer Center (MSKCC) and Gene Expression Omnibus (GEO) datasets, as well as in this study unit cohort. The genes were examined via functional enrichment analysis, and the tumor immune features, tumor mutation features and copy number variations (CNVs) of patients with different risk scores were analysed. The response of patients to multiple chemotherapeutic/targeted drugs was assessed using the pRRophetic algorithm, and immunotherapy was inferred by the Tumor Immune Dysfunction and Exclusion (TIDE) and immunophenoscore (IPS). Results Cuproptosis-related immune risk scores (CRIRSs) were developed based on PRLR, DES and LECT2. High CRIRSs indicated poor overall survival (OS), disease-free survival (DFS) in the TCGA-PRAD, MSKCC and GEO datasets and higher T stage and Gleason scores in TCGA-PRAD. Similarly, in the sample collected by the study unit, patients with high CRIRS had higher T-stage and Gleason scores. Additionally, higher CRIRSs were negatively correlated with the abundance of activated B cells, activated CD8+ T cells and other stromal or immune cells. The expression of some immune checkpoints was negatively correlated with CRIRSs. Tumor mutational burden (TMB), mutant-allele tumor heterogeneity (MATH) and copy number variation (CNV) scores were all higher in the high-CRIRS group. Multiple chemotherapeutic/targeted drugs and immunotherapy had better responsiveness in the low-CRIRS group. Conclusion Overall, lower CRIRS indicated better response to treatment strategies and better prognostic outcomes.
Collapse
Affiliation(s)
- Kai Yao
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rumeng Zhang
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Liang Li
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingdong Liu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shiyao Feng
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haixin Yan
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihui Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dongdong Xie
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Urology, Affiliated Fuyang Hospital of Anhui Medical University, Fuyang, Anhui, China
| |
Collapse
|
2
|
Khatami F, Hassanzad M, Nikfar S, Guitynavard F, Karimaee S, Tamehri Zadeh SS, Gholami K, Rezaeian A, Feiz-Abadi SA, Jahanshahi F, Aghamir SMK. The importance of personalized medicine in urological cancers. J Diabetes Metab Disord 2022; 21:841-852. [PMID: 35673449 PMCID: PMC9167380 DOI: 10.1007/s40200-021-00824-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 01/10/2023]
Abstract
A better understanding of key regulatory pathways involved in cancers has led to the development of molecularly targeted therapies. Molecular profiling based on genomics, proteomics, and metabolomics in tumors provides clinicians with the necessary information to maintain a personalized therapeutic regimen according to the patient's needs. for example, androgen deprivation therapy (ADT) for advanced prostate cancer is one of the earliest forms of targeted therapy and has remained a choice of treatment by physicians. Unfortunately, most patients will eventually become non-responsive to ADT and succumb to the disease. Since the emergence of ADT, the understanding of androgen receptor (AR) signaling and mechanisms driving the resistance to ADT has been significantly improved. Inactivation of the PTEN gene is a common occurrence in prostate cancers and is associated with metastatic potential, androgen independence, and poor prognosis. Several studies over personalized medicine for muscle-invasive and metastatic bladder cancer discussed potential molecular biomarkers which are currently under investigation and based on the excision repair cross-complementing group 1 (ERCC1) gene and its role in tumor development and therapeutic resistance to cytotoxic DNA-damaging chemotherapy and ionizing radiation. In this review, we consider personalized medicine for four urological cancers.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Hassanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shekoufeh Nikfar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Guitynavard
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Karimaee
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Keykavos Gholami
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - AhmadReza Rezaeian
- Department of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
3
|
Kurose H, Ueda K, Ogasawara N, Chikui K, Nakiri M, Nishihara K, Matsuo M, Suekane S, Kusano H, Akiba J, Yano H, Igawa T. Impact of Gleason score of the tumor at the positive surgical margin as a prognostic factor. Mol Clin Oncol 2022; 16:82. [PMID: 35251633 PMCID: PMC8892462 DOI: 10.3892/mco.2022.2515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/27/2021] [Indexed: 11/19/2022] Open
Abstract
Although numerous studies have reported that a positive surgical margin (PSM) is the most important predictive factor for biochemical recurrence (BCR) of prostate cancer (PCa), only a small number of studies have evaluated the predictive value of the Gleason score (GS) of the tumor at the margin in radical prostatectomy (RP). The present study aimed to investigate the preoperative factors that predict PSM and the significant predictive factors for BCR in cases with PSM. In addition, it was examined whether documenting the GS of the tumor at the margin in pathological reports is useful as a predictive factor for BCR. Data of 241 patients with PCa who underwent RP at Kurume University Hospital (Kurume, Japan) between January 2007 and December 2011 were retrospectively reviewed. The median follow-up period was 72 months and 122 patients had at least one PSM. The time to BCR was significantly shorter in patients with PSM than in those with a negative surgical margin. Multivariate analysis demonstrated that >10 ng/ml prostate-specific antigen at diagnosis (P=0.024) and >25% positive core at biopsy (P=0.041) were independent prognostic preoperative factors for PSM. The GS of the tumor at the margin was equal, lower and higher than those of the main tumor in 74 (60.7%), 16 (13.1%) and 32 (26.2%) RPs, respectively. The BCR rates were 35.7, 55.1 and 82.1% in patients whose GS of the tumor at the margin was 6, 7 and 8-10, respectively (P=0.0017). The GS of the tumor at the PSM (P=0.038) and anatomic location of the PSM (P=0.04) were identified as independent prognostic preoperative factors for BCR, whereas the GS of the main tumor and margin length were not. These results suggest that documenting the GS at the margin in pathological reports is useful as a predictive factor for BCR.
Collapse
Affiliation(s)
- Hirofumi Kurose
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Kosuke Ueda
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Naoyuki Ogasawara
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Katsuaki Chikui
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Makoto Nakiri
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Kiyoaki Nishihara
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Mitsunori Matsuo
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Hironori Kusano
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Jun Akiba
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| |
Collapse
|
4
|
Singh J, Sohal SS, Ahuja K, Lim A, Duncan H, Thachil T, De Ieso P. Investigation of circulatory cytokines in patients undergoing intensity-modulated radiotherapy (IMRT) for adenocarcinoma of the prostate and association with acute RT-induced toxicity: A prospective clinical study. Cytokine 2020; 131:155108. [PMID: 32330791 DOI: 10.1016/j.cyto.2020.155108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/29/2020] [Accepted: 04/17/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Jagtar Singh
- College of Health and Human Sciences, Charles Darwin University, Northern Territory, Australia.
| | | | - Kiran Ahuja
- School of Health Sciences, University of Tasmania, Tasmania, Australia
| | - Aijye Lim
- Department of Anatomical Pathology, Royal Darwin Hospital, Northern Territory, Australia.
| | - Henry Duncan
- Darwin Private Hospital, Royal Darwin Hospital, Northern Territory, Australia.
| | - Thanuja Thachil
- Austin Radiation Oncology Centre, Ballarat, Victoria, Australia.
| | - Paolo De Ieso
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| |
Collapse
|
5
|
KPNA2/ERG Coexpression is Associated With Early Recurrence in Advanced Prostate Cancers. Appl Immunohistochem Mol Morphol 2019; 28:62-66. [PMID: 31809312 DOI: 10.1097/pai.0000000000000706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Most prostate cancers (PC) overexpress the ERG oncogene and karyopherin α 2 (KPNA2). These genes play a role in prostatic carcinogenesis, but their prognostic significance is still debated. The aim of this study was to determine the prognostic significance of ERG and KPNA2 expression, and their association to early prostate-specific antigen (PSA) biochemical recurrence in advanced PC with lymph node metastases. A series of 65 consecutive pN1 M0 R0 PC samples obtained by radical prostatectomy with lymphadenectomy has been analyzed for ERG and KPNA2 expression by immunohistochemistry. For each case, the following clinical data were collected: age, preoperative serum PSA levels, Gleason grade group, TNM stage, and follow-up. PC recurrence was investigated by serum PSA assay and defined by a PSA concentration >0.2 ng/mL after a nadir of <0.1 ng/mL following radical prostatectomy. ERG-positive staining was found in 25/65 cases (38%), and KPNA2 in 56/65 cases (86%); neither was detected in normal prostatic tissue. Immunohistochemical concordance was found between primary tumor and lymph node metastases in 24/25 (96%) of ERG and 53/56 (95%) of KPNA2-positive cases. The follow-up was known in all cases, and early PSA recurrence occurred in 25/65 cases (38%). ERG positivity, both alone and in conjunction with KPNA2 positivity, was strongly associated with early PSA recurrence [both ERG+ and KPNA+, odds ratio: 22.2 (95% confidence interval, 6.0-82.3); ERG+ alone odds ratio: 17.9 (95% confidence interval, 5.1-63.5); P<0.0001 for both]. KPNA2 expression was significantly associated with the tumor stage (P<0.00001). The results suggest that the ERG+ phenotype might be selected in metastasis-initiating clones. ERG and KPNA2 may have a prognostic value, and their positivity in PC might warrant more aggressive treatments.
Collapse
|
6
|
Becker F, Joerg V, Hupe MC, Roth D, Krupar R, Lubczyk V, Kuefer R, Sailer V, Duensing S, Kirfel J, Merseburger AS, Brägelmann J, Perner S, Offermann A. Increased mediator complex subunit CDK19 expression associates with aggressive prostate cancer. Int J Cancer 2019; 146:577-588. [PMID: 31271443 DOI: 10.1002/ijc.32551] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 12/23/2022]
Abstract
The Mediator complex is a transcriptional regulator interacting with transcription factors and RNA-polymerase-II. Recently, we identified its subunit CDK19 to be specifically expressed in prostate cancer (PCa) and to be functionally implicated in PCa aggressiveness. Aim of our study was to comprehensively characterize the protein expression of CDK19 and its paralog CDK8 in PCa. We performed immunohistochemistry (IHC) for CDK19/CDK8 on a large cohort including needle biopsies from 202 patients, 799 primary tumor foci of radical prostatectomy specimens from 415 patients, 120 locally advanced tumor foci obtained by palliative transurethral resection, 140 lymph node metastases, 67 distant metastases and 82 benigns. Primary tumors were stained for the proliferation marker Ki67, androgen receptor (AR) and ERG. For 376 patients, clinic-pathologic data were available. Primary endpoint was disease-recurrence-free survival (DFS). Nuclear CDK19 and CDK8 expression increases during progression showing the highest intensity in metastatic and castration-resistant tumors. High CDK19 expression on primary tumors correlates with DFS independently from Gleason grade and PSA. Five-year-DFS rates of patients with primary tumors expressing no, moderate and high CDK19 are 73.7, 56.9 and 30.4%, respectively. CDK19 correlates with Gleason grade, T-stage, Ki67 proliferation-index, nuclear AR expression and ERG-status. Therapeutic options for metastatic and castration-resistant PCa remain limited. In the current study, we confirmed an important role of the Mediator subunit CDK19 in advanced PCa supporting current developments to target CDK19 and its paralog CDK8. Furthermore, CDK19 protein expression has the potential to predict disease recurrence independently from established biomarkers thus contributing to individual management for PCa patients.
Collapse
Affiliation(s)
- Finn Becker
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Vincent Joerg
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Marie C Hupe
- Department of Urology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Doris Roth
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | | | - Verena Lubczyk
- Department of Pathology, Klinik am Eichert Alb Fils Kliniken, Goeppingen, Germany
| | - Rainer Kuefer
- Department of Urology, Klinik am Eichert Alb Fils Kliniken, Goeppingen, Germany
| | - Verena Sailer
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Jutta Kirfel
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Johannes Brägelmann
- Molecular Pathology, Institute of Pathology, University Hospital of Cologne, Cologne, Germany.,Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne, Germany
| | - Sven Perner
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Anne Offermann
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| |
Collapse
|
7
|
Kurose H, Naito Y, Akiba J, Kondo R, Ogasawara S, Kusano H, Sanada S, Abe H, Kakuma T, Ueda K, Igawa T, Yano H. High sulfite oxidase expression could predict postoperative biochemical recurrence in patients with prostate cancer. Med Mol Morphol 2019; 52:164-172. [PMID: 30631948 DOI: 10.1007/s00795-018-00214-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Abstract
Sulfite oxidase (SUOX) is a metalloenzyme that plays a role in ATP synthesis via oxidative phosphorylation in mitochondria and has been reported to also be involved in the invasion and differentiation capacities of tumor cells. Here, we performed a clinicopathological investigation of SUOX expression in prostate cancer and discussed the usefulness of SUOX expression as a predictor of biochemical recurrence following surgical treatment in prostate cancer. This study was conducted using Tissue Micro Array specimens obtained from 97 patients who underwent radical prostatectomy at our hospital between 2007 and 2011. SUOX staining was used to evaluate cytoplasmic SUOX expression. In the high-expression group, the early biochemical recurrence was significantly more frequent than in the low-expression group (p = 0.0008). In multivariate analysis, high SUOX expression was found to serve as an independent prognostic factor of biochemical recurrence (hazard ratio = 2.33, 95% confidence interval = 1.32-4.15, p = 0.0037). In addition, Ki-67-labeling indices were significantly higher in the high-expression group than in the low-expression group (p = 0.0058). Therefore, SUOX expression may be a powerful prognostic biomarker for decision-making in postoperative follow-up after total prostatectomy and with regard to the need for relief treatment.
Collapse
Affiliation(s)
- Hirofumi Kurose
- Department of Pathology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.,Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Yoshiki Naito
- Department of Pathology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan. .,Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan.
| | - Jun Akiba
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan
| | - Reiichiro Kondo
- Department of Pathology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Sachiko Ogasawara
- Department of Pathology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Hironori Kusano
- Department of Pathology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Sakiko Sanada
- Department of Pathology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Hideyuki Abe
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan
| | - Tatsuyuki Kakuma
- Biostatistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Kosuke Ueda
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| |
Collapse
|
8
|
Yang F, Ford JC, Dogan N, Padgett KR, Breto AL, Abramowitz MC, Dal Pra A, Pollack A, Stoyanova R. Magnetic resonance imaging (MRI)-based radiomics for prostate cancer radiotherapy. Transl Androl Urol 2018; 7:445-458. [PMID: 30050803 PMCID: PMC6043736 DOI: 10.21037/tau.2018.06.05] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 06/05/2018] [Indexed: 11/25/2022] Open
Abstract
In radiotherapy (RT) of prostate cancer, dose escalation has been shown to reduce biochemical failure. Dose escalation only to determinate prostate tumor habitats has the potential to improve tumor control with less toxicity than when the entire prostate is dose escalated. Other issues in the treatment of the RT patient include the choice of the RT technique (hypo- or standard fractionation) and the use and length of concurrent/adjuvant androgen deprivation therapy (ADT). Up to 50% of high-risk men demonstrate biochemical failure suggesting that additional strategies for defining and treating patients based on improved risk stratification are required. The use of multiparametric MRI (mpMRI) is rapidly gaining momentum in the management of prostate cancer because of its improved diagnostic potential and its ability to combine functional and anatomical information. Currently, the Prostate Imaging, Reporting and Diagnosis System (PIRADS) is the standard of care for region of interest (ROI) identification and risk classification. However, PIRADS was not designed for 3D tumor volume delineation; there is a large degree of subjectivity and PIRADS does not accurately and reproducibly elucidate inter- and intra-lesional spatial heterogeneity. "Radiomics", as it refers to the extraction and analysis of large number of advanced quantitative radiological features from medical images using high throughput methods, is perfectly suited as an engine to effectively sift through the multiple series of prostate mpMRI sequences and quantify regions of interest. The radiomic efforts can be summarized in two main areas: (I) detection/segmentation of the suspicious lesion; and (II) assessment of the aggressiveness of prostate cancer. As related to RT, the goal of the latter is in particular to identify patients at high risk for metastatic disease; and the aim of the former is to identify and segment cancerous lesions and thus provide targets for radiation boost. The article is structured as follows: first, we describe the radiomic approach; and second, we discuss the radiomic pipeline as tailored for RT of prostate cancer. In this process we summarize the current efforts and progress in integrating mpMRI radiomics into the radiotherapeutic management of prostate cancer with emphasis placed on its role in treatment target definition, treatment plan strategizing, and prognostic assessment. The described concepts, methods and tools are not currently applicable to the radiation oncology practice outside of the research setting. More data are required in the form of clinical trials to assess the robustness of radiomics-based predictive models, and to maximize the efficacy of these models.
Collapse
Affiliation(s)
- Fei Yang
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - John C. Ford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Nesrin Dogan
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kyle R. Padgett
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Adrian L. Breto
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matthew C. Abramowitz
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
9
|
18F-Fluorocholine PET Whole-Body MRI in the Staging of High-Risk Prostate Cancer. AJR Am J Roentgenol 2018; 210:635-640. [PMID: 29323548 DOI: 10.2214/ajr.17.18567] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The purpose of this study was to determine whether integrated 18F-fluorocholine (FCH) PET whole-body MRI (PET/WBMRI) depicts lymph node and distant metastases in patients with high-risk prostate cancer more frequently than does conventional staging. SUBJECTS AND METHODS A prospective study included 58 patients with untreated high-risk prostate cancer. After conventional staging (CT and bone scintigraphy), patients underwent FCH PET/WBMRI (n = 10) or FCH PET/CT and WBMRI (n = 48). Metastatic sites and disease stage were recorded for each modality (conventional imaging, PET, WBMRI, and PET/WBMRI) and compared with a standard of reference (histopathologic examination, imaging, and clinical follow-up) and early clinical outcomes. RESULTS In the detection of metastases, PET had significantly higher sensitivity (72/77 [93.5%]) than conventional imaging (49/77 [63.6%]; p < 0.001) and WBMRI (56/77 [72.7%]; p = 0.002). There was a trend toward improved detection with PET/WBMRI (77/77 [100%]) compared with PET alone (p = 0.059). For correct NM staging, PET and PET/WBMRI performed better than conventional imaging (p = 0.002) and WBMRI (p = 0.008). Twelve of 56 patients (21.4%) had early biochemical failure after radical treatment (median, 7 months; range, 1-20 months). This rate was higher for patients with M1a or M1b disease at PET/WBMRI than for others, but this finding did not reach statistical significance (4/8 [50%] vs 8/48 [16.7%]; p = 0.055). CONCLUSION In patients with high-risk prostate cancer, FCH PET and FCH PET/WBMRI depict significantly more metastatic lesions than do conventional imaging and WBMRI. Stage determined with PET/WBMRI may correlate with early outcomes.
Collapse
|
10
|
Chaiswing L, Weiss HL, Jayswal RD, St. Clair DK, Kyprianou N. Profiles of Radioresistance Mechanisms in Prostate Cancer. Crit Rev Oncog 2018; 23:39-67. [PMID: 29953367 PMCID: PMC6231577 DOI: 10.1615/critrevoncog.2018025946] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Radiation therapy (RT) is commonly used for the treatment of localized prostate cancer (PCa). However, cancer cells often develop resistance to radiation through unknown mechanisms and pose an intractable challenge. Radiation resistance is highly unpredictable, rendering the treatment less effective in many patients and frequently causing metastasis and cancer recurrence. Understanding the molecular events that cause radioresistance in PCa will enable us to develop adjuvant treatments for enhancing the efficacy of RT. Radioresistant PCa depends on the elevated DNA repair system and the intracellular levels of reactive oxygen species (ROS) to proliferate, self-renew, and scavenge anti-cancer regimens, whereas the elevated heat shock protein 90 (HSP90) and the epithelial-mesenchymal transition (EMT) enable radioresistant PCa cells to metastasize after exposure to radiation. The up-regulation of the DNA repairing system, ROS, HSP90, and EMT effectors has been studied extensively, but not targeted by adjuvant therapy of radioresistant PCa. Here, we emphasize the effects of ionizing radiation and the mechanisms driving the emergence of radioresistant PCa. We also address the markers of radioresistance, the gene signatures for the predictive response to radiotherapy, and novel therapeutic platforms for targeting radioresistant PCa. This review provides significant insights into enhancing the current knowledge and the understanding toward optimization of these markers for the treatment of radioresistant PCa.
Collapse
Affiliation(s)
| | - Heidi L. Weiss
- The Markey Biostatistics and Bioinformatics Shared Resource Facility
| | - Rani D. Jayswal
- The Markey Biostatistics and Bioinformatics Shared Resource Facility
| | | | - Natasha Kyprianou
- Department of Toxicology and Cancer Biology
- Department of Urology
- Department of Biochemistry, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
11
|
Translating a Prognostic DNA Genomic Classifier into the Clinic: Retrospective Validation in 563 Localized Prostate Tumors. Eur Urol 2017; 72:22-31. [DOI: 10.1016/j.eururo.2016.10.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/10/2016] [Indexed: 01/27/2023]
|
12
|
Evaluation of inflammatory cytokines and oxidative stress markers in prostate cancer patients undergoing curative radiotherapy. Cent Eur J Immunol 2017; 42:68-72. [PMID: 28680332 PMCID: PMC5470615 DOI: 10.5114/ceji.2017.67319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/20/2016] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer is the second leading cause of cancer-related death in men. The present study was carried out to investigate the radiation response of serum cytokines and oxidative markers to find out if these novel biomarkers have significant applications regarding radiation outcome in prostate cancer patients. Significant elevations of prostatic specific antigen (PSA), asymmetric dimethyl arginine (ADMA) and nitric oxide (NO) were recorded in cancer prostate patients at the time of diagnosis compared to controls. Patients were subjected to radiotherapy post prostatectomy with a total dose of 66 Gy in 33 fractions (5 sessions/week) for 7 weeks. At the end of the seventh week post radiotherapy, ADMA levels were accentuated while the levels of PSA and NO were lower than before therapy. The level of inflammatory cytokines (interleukins IL-4, IL-5 and interferon-gamma) in post radiation therapy patients were significantly elevated compared to both controls and prostate cancer patients. A significant inverse correlation was observed in prostate cancer patients between ADMA and NO. Moreover, a significant inverse correlation in post radiation therapy patients was observed between IL-5 and PSA. These results are highly suggestive that there is a specific cytokine response in patients undergoing curative radiotherapy for prostate cancer.
Collapse
|
13
|
Stoyanova R, Takhar M, Tschudi Y, Ford JC, Solórzano G, Erho N, Balagurunathan Y, Punnen S, Davicioni E, Gillies RJ, Pollack A. Prostate cancer radiomics and the promise of radiogenomics. Transl Cancer Res 2016; 5:432-447. [PMID: 29188191 DOI: 10.21037/tcr.2016.06.20] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostate cancer exhibits intra-tumoral heterogeneity that we hypothesize to be the leading confounding factor contributing to the underperformance of the current pre-treatment clinical-pathological and genomic assessment. These limitations impose an urgent need to develop better computational tools to identify men with low risk of prostate cancer versus others that may be at risk for developing metastatic cancer. The patient stratification will directly translate to patient treatments, wherein decisions regarding active surveillance or intensified therapy are made. Multiparametric MRI (mpMRI) provides the platform to investigate tumor heterogeneity by mapping the individual tumor habitats. We hypothesize that quantitative assessment (radiomics) of these habitats results in distinct combinations of descriptors that reveal regions with different physiologies and phenotypes. Radiogenomics, a discipline connecting tumor morphology described by radiomic and its genome described by the genomic data, has the potential to derive "radio phenotypes" that both correlate to and complement existing validated genomic risk stratification biomarkers. In this article we first describe the radiomic pipeline, tailored for analysis of prostate mpMRI, and in the process we introduce our particular implementations of radiomics modules. We also summarize the efforts in the radiomics field related to prostate cancer diagnosis and assessment of aggressiveness. Finally, we describe our results from radiogenomic analysis, based on mpMRI-Ultrasound (MRI-US) biopsies and discuss the potential of future applications of this technique. The mpMRI radiomics data indicate that the platform would significantly improve the biopsy targeting of prostate habitats through better recognition of indolent versus aggressive disease, thereby facilitating a more personalized approach to prostate cancer management. The expectation to non-invasively identify habitats with high probability of housing aggressive cancers would result in directed biopsies that are more informative and actionable. Conversely, providing evidence for lack of disease would reduce the incidence of non-informative biopsies. In radiotherapy of prostate cancer, dose escalation has been shown to reduce biochemical failure. Dose escalation only to determinate prostate habitats has the potential to improve tumor control with less toxicity than when the entire prostate is dose escalated.
Collapse
Affiliation(s)
- Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mandeep Takhar
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | - Yohann Tschudi
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - John C Ford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gabriel Solórzano
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicholas Erho
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | | | - Sanoj Punnen
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elai Davicioni
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | - Robert J Gillies
- Cancer Imaging and Metabolism, Moffitt Cancer Center, Tampa, FL, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
14
|
Theophilou G, Lima KMG, Briggs M, Martin-Hirsch PL, Stringfellow HF, Martin FL. A biospectroscopic analysis of human prostate tissue obtained from different time periods points to a trans-generational alteration in spectral phenotype. Sci Rep 2015; 5:13465. [PMID: 26310632 PMCID: PMC4550877 DOI: 10.1038/srep13465] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/28/2015] [Indexed: 02/04/2023] Open
Abstract
Prostate cancer is the most commonly-diagnosed malignancy in males worldwide; however, there is marked geographic variation in incidence that may be associated with a Westernised lifestyle. We set out to determine whether attenuated total reflection Fourier-transform infrared (ATR-FTIR) or Raman spectroscopy combined with principal component analysis-linear discriminant analysis or variable selection techniques employing genetic algorithm or successive projection algorithm could be utilised to explore differences between prostate tissues from differing years. In total, 156 prostate tissues from transurethral resection of the prostate procedures for benign prostatic hyperplasia from 1983 to 2013 were collected. These were distributed to form seven categories: 1983–1984 (n = 20), 1988–1989 (n = 25), 1993–1994 (n = 21), 1998–1999 (n = 21), 2003–2004 (n = 21), 2008–2009 (n = 20) and 2012–2013 (n = 21). Ten-μm-thick tissue sections were floated onto Low-E (IR-reflective) slides for ATR-FTIR or Raman spectroscopy. The prostate tissue spectral phenotype altered in a temporal fashion. Examination of the two categories that are at least one generation (30 years) apart indicated highly-significant segregation, especially in spectral regions containing DNA and RNA bands (≈1,000–1,490 cm−1). This may point towards alterations that have occurred through genotoxicity or through epigenetic modifications. Immunohistochemical studies for global DNA methylation supported this. This study points to a trans-generational phenotypic change in human prostate.
Collapse
Affiliation(s)
- Georgios Theophilou
- Centre for Biophotonics, LEC, Lancaster University, Lancaster LA1 4YQ, UK.,Department of Obstetrics and Gynaecology, Central Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Kássio M G Lima
- Centre for Biophotonics, LEC, Lancaster University, Lancaster LA1 4YQ, UK.,Institute of Chemistry, Biological Chemistry and Chemometrics, Federal University of Rio Grande do Norte, Natal 59072-970, RN-Brazil
| | - Matthew Briggs
- Department of Obstetrics and Gynaecology, Central Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Pierre L Martin-Hirsch
- Centre for Biophotonics, LEC, Lancaster University, Lancaster LA1 4YQ, UK.,Department of Obstetrics and Gynaecology, Central Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Helen F Stringfellow
- Department of Obstetrics and Gynaecology, Central Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Francis L Martin
- Centre for Biophotonics, LEC, Lancaster University, Lancaster LA1 4YQ, UK
| |
Collapse
|
15
|
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: 2.9] [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
|
16
|
Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, Meng A, Hennings-Yeomans PH, McPherson A, Sabelnykova VY, Zia A, Fox NS, Livingstone J, Shiah YJ, Wang J, Beck TA, Have CL, Chong T, Sam M, Johns J, Timms L, Buchner N, Wong A, Watson JD, Simmons TT, P'ng C, Zafarana G, Nguyen F, Luo X, Chu KC, Prokopec SD, Sykes J, Dal Pra A, Berlin A, Brown A, Chan-Seng-Yue MA, Yousif F, Denroche RE, Chong LC, Chen GM, Jung E, Fung C, Starmans MHW, Chen H, Govind SK, Hawley J, D'Costa A, Pintilie M, Waggott D, Hach F, Lambin P, Muthuswamy LB, Cooper C, Eeles R, Neal D, Tetu B, Sahinalp C, Stein LD, Fleshner N, Shah SP, Collins CC, Hudson TJ, McPherson JD, van der Kwast T, Bristow RG. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat Genet 2015; 47:736-45. [PMID: 26005866 DOI: 10.1038/ng.3315] [Citation(s) in RCA: 351] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/01/2015] [Indexed: 12/12/2022]
Abstract
Herein we provide a detailed molecular analysis of the spatial heterogeneity of clinically localized, multifocal prostate cancer to delineate new oncogenes or tumor suppressors. We initially determined the copy number aberration (CNA) profiles of 74 patients with index tumors of Gleason score 7. Of these, 5 patients were subjected to whole-genome sequencing using DNA quantities achievable in diagnostic biopsies, with detailed spatial sampling of 23 distinct tumor regions to assess intraprostatic heterogeneity in focal genomics. Multifocal tumors are highly heterogeneous for single-nucleotide variants (SNVs), CNAs and genomic rearrangements. We identified and validated a new recurrent amplification of MYCL, which is associated with TP53 deletion and unique profiles of DNA damage and transcriptional dysregulation. Moreover, we demonstrate divergent tumor evolution in multifocal cancer and, in some cases, tumors of independent clonal origin. These data represent the first systematic relation of intraprostatic genomic heterogeneity to predicted clinical outcome and inform the development of novel biomarkers that reflect individual prognosis.
Collapse
Affiliation(s)
- Paul C Boutros
- 1] Ontario Institute for Cancer Research, Toronto, Ontario, Canada. [2] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. [3] Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Fraser
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | | | - Dominique Trudel
- Department of Pathology and Laboratory Medicine, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Emilie Lalonde
- 1] Ontario Institute for Cancer Research, Toronto, Ontario, Canada. [2] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Alice Meng
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | | | - Andrew McPherson
- School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Amin Zia
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Natalie S Fox
- 1] Ontario Institute for Cancer Research, Toronto, Ontario, Canada. [2] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Yu-Jia Shiah
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jianxin Wang
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Timothy A Beck
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Cherry L Have
- Department of Pathology and Laboratory Medicine, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Taryne Chong
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Michelle Sam
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jeremy Johns
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Lee Timms
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Ada Wong
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - John D Watson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Trent T Simmons
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Christine P'ng
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gaetano Zafarana
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Francis Nguyen
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Xuemei Luo
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Kenneth C Chu
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Jenna Sykes
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Alejandro Berlin
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Brown
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Fouad Yousif
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Lauren C Chong
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gregory M Chen
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Esther Jung
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Clement Fung
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Hanbo Chen
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - James Hawley
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Alister D'Costa
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Melania Pintilie
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Daryl Waggott
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Faraz Hach
- School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Philippe Lambin
- Department of Radiotherapy, Maastricht University, Maastricht, the Netherlands
| | | | - Colin Cooper
- 1] Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK. [2] Department of Biological Sciences, University of East Anglia, Norwich, UK. [3] School of Medicine, University of East Anglia, Norwich, UK
| | - Rosalind Eeles
- 1] Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK. [2] Royal Marsden National Health Service (NHS) Foundation Trust, London and Sutton, UK
| | - David Neal
- 1] Urological Research Laboratory, Cancer Research UK Cambridge Research Institute, Cambridge, UK. [2] Department of Surgical Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Bernard Tetu
- Department of Pathology, Laval University, Quebec City, Quebec, Canada
| | - Cenk Sahinalp
- School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Lincoln D Stein
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Neil Fleshner
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sohrab P Shah
- 1] Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada. [2] Department of Computer Science, University of British Columbia, Vancouver, British Columbia, Canada. [3] British Columbia Cancer Agency Research Centre, Vancouver, British Columbia, Canada
| | - Colin C Collins
- 1] Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada. [2] Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Thomas J Hudson
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Theodorus van der Kwast
- Department of Pathology and Laboratory Medicine, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Robert G Bristow
- 1] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. [2] Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. [3] Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Locke JA, Dal Pra A, Supiot S, Warde P, Bristow RG. Synergistic action of image-guided radiotherapy and androgen deprivation therapy. Nat Rev Urol 2015; 12:193-204. [PMID: 25800395 DOI: 10.1038/nrurol.2015.50] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The combined use of androgen deprivation therapy (ADT) and image-guided radiotherapy (IGRT) can improve overall survival in aggressive, localized prostate cancer. However, owing to the adverse effects of prolonged ADT, it is imperative to identify the patients who would benefit from this combined-modality therapy relative to the use of IGRT alone. Opportunities exist for more personalized approaches in treating aggressive, locally advanced prostate cancer. Biomarkers--such as disseminated tumour cells, circulating tumour cells, genomic signatures and molecular imaging techniques--could identify the patients who are at greatest risk for systemic metastases and who would benefit from the addition of systemic ADT. By contrast, when biomarkers of systemic disease are not present, treatment could proceed using local IGRT alone. The choice of drug, treatment duration and timing of ADT relative to IGRT could be predicated on these personalized approaches to prostate cancer medicine. These novel treatment intensification and reduction strategies could result in improved prostate-cancer-specific survival and overall survival, without incurring the added expense of metabolic syndrome and other adverse effects of ADT in all patients.
Collapse
Affiliation(s)
- Jennifer A Locke
- Department of Urologic Sciences, University of British Columbia, Gordon &Leslie Diamond Health Care Centre, Level 6, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada
| | - Alan Dal Pra
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 4, CH-3010 Bern, Switzerland
| | - Stéphane Supiot
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes-St-Herblain, 8 quai Moncousu, BP 70721, 44000 Nantes, France
| | - Padraig Warde
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - Robert G Bristow
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
18
|
Liu M, Yao XD, Li W, Geng J, Yan Y, Che JP, Xu YF, Zheng JH. Nrf2 sensitizes prostate cancer cells to radiation via decreasing basal ROS levels. Biofactors 2015; 41:52-7. [PMID: 25728635 DOI: 10.1002/biof.1200] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/19/2015] [Accepted: 01/20/2015] [Indexed: 11/07/2022]
Abstract
Androgen deprivation therapy (ADT) was reported to lower basal ROS level in prostate cancer (PCa) and to sensitize PCa to radiation. We aimed to seek for the underlying molecular mechanism and to develop novel additive treatments to ADT in this regard. We simulated human androgen milieu in vitro and tested the ROS level in PCa cells undergoing ADT. We also tested the Nrf2 level in PCa cells with or without ADT. Genetic and pharmaceutical upregulation of Nrf2 was applied in vitro and in vivo in transgenic adenocarcinoma of the mouse prostate (TRAMP) mice with or without castration to investigate whether Nrf2 overexpression supplemented the effect of ADT in PCa. We first discovered that androgen deprivation increased basal ROS level in PCa cells with AR expression. We then found that genetic Nrf2 upregulation lowered basal ROS similar to ADT. Also, SFN sensitized PCa cell to radiation via upregulation of Nrf2. We then found that Nrf2 level in control TRAMP groups was lower than castration or SFN groups. The SFN treated TRAMP mice showed similar level of Nrf2 to castration. Genetic and pharmaceutical upregulation of Nrf2 lowered the ROS in PCa cells and sensitized PCa cells to radiation similar to ADT, implicating possible administration of SFN in place of ADT for PCa patients requiring radiotherapy.
Collapse
Affiliation(s)
- Min Liu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Lalonde E, Ishkanian AS, Sykes J, Fraser M, Ross-Adams H, Erho N, Dunning MJ, Halim S, Lamb AD, Moon NC, Zafarana G, Warren AY, Meng X, Thoms J, Grzadkowski MR, Berlin A, Have CL, Ramnarine VR, Yao CQ, Malloff CA, Lam LL, Xie H, Harding NJ, Mak DYF, Chu KC, Chong LC, Sendorek DH, P'ng C, Collins CC, Squire JA, Jurisica I, Cooper C, Eeles R, Pintilie M, Dal Pra A, Davicioni E, Lam WL, Milosevic M, Neal DE, van der Kwast T, Boutros PC, Bristow RG. Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort study. Lancet Oncol 2014; 15:1521-1532. [PMID: 25456371 DOI: 10.1016/s1470-2045(14)71021-6] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Clinical prognostic groupings for localised prostate cancers are imprecise, with 30-50% of patients recurring after image-guided radiotherapy or radical prostatectomy. We aimed to test combined genomic and microenvironmental indices in prostate cancer to improve risk stratification and complement clinical prognostic factors. METHODS We used DNA-based indices alone or in combination with intra-prostatic hypoxia measurements to develop four prognostic indices in 126 low-risk to intermediate-risk patients (Toronto cohort) who will receive image-guided radiotherapy. We validated these indices in two independent cohorts of 154 (Memorial Sloan Kettering Cancer Center cohort [MSKCC] cohort) and 117 (Cambridge cohort) radical prostatectomy specimens from low-risk to high-risk patients. We applied unsupervised and supervised machine learning techniques to the copy-number profiles of 126 pre-image-guided radiotherapy diagnostic biopsies to develop prognostic signatures. Our primary endpoint was the development of a set of prognostic measures capable of stratifying patients for risk of biochemical relapse 5 years after primary treatment. FINDINGS Biochemical relapse was associated with indices of tumour hypoxia, genomic instability, and genomic subtypes based on multivariate analyses. We identified four genomic subtypes for prostate cancer, which had different 5-year biochemical relapse-free survival. Genomic instability is prognostic for relapse in both image-guided radiotherapy (multivariate analysis hazard ratio [HR] 4·5 [95% CI 2·1-9·8]; p=0·00013; area under the receiver operator curve [AUC] 0·70 [95% CI 0·65-0·76]) and radical prostatectomy (4·0 [1·6-9·7]; p=0·0024; AUC 0·57 [0·52-0·61]) patients with prostate cancer, and its effect is magnified by intratumoral hypoxia (3·8 [1·2-12]; p=0·019; AUC 0·67 [0·61-0·73]). A novel 100-loci DNA signature accurately classified treatment outcome in the MSKCC low-risk to intermediate-risk cohort (multivariate analysis HR 6·1 [95% CI 2·0-19]; p=0·0015; AUC 0·74 [95% CI 0·65-0·83]). In the independent MSKCC and Cambridge cohorts, this signature identified low-risk to high-risk patients who were most likely to fail treatment within 18 months (combined cohorts multivariate analysis HR 2·9 [95% CI 1·4-6·0]; p=0·0039; AUC 0·68 [95% CI 0·63-0·73]), and was better at predicting biochemical relapse than 23 previously published RNA signatures. INTERPRETATION This is the first study of cancer outcome to integrate DNA-based and microenvironment-based failure indices to predict patient outcome. Patients exhibiting these aggressive features after biopsy should be entered into treatment intensification trials. FUNDING Movember Foundation, Prostate Cancer Canada, Ontario Institute for Cancer Research, Canadian Institute for Health Research, NIHR Cambridge Biomedical Research Centre, The University of Cambridge, Cancer Research UK, Cambridge Cancer Charity, Prostate Cancer UK, Hutchison Whampoa Limited, Terry Fox Research Institute, Princess Margaret Cancer Centre Foundation, PMH-Radiation Medicine Program Academic Enrichment Fund, Motorcycle Ride for Dad (Durham), Canadian Cancer Society.
Collapse
Affiliation(s)
- Emilie Lalonde
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Adrian S Ishkanian
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, USA
| | - Jenna Sykes
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michael Fraser
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Helen Ross-Adams
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Nicholas Erho
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | - Mark J Dunning
- Bioinformatics Core, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Silvia Halim
- Bioinformatics Core, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Alastair D Lamb
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Department of Urology, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Nathalie C Moon
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Gaetano Zafarana
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Anne Y Warren
- Department of Pathology, Addenbrooke's Hospital, Cambridge, UK
| | - Xianyue Meng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - John Thoms
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michal R Grzadkowski
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Alejandro Berlin
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Cherry L Have
- Department of Pathology, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - Varune R Ramnarine
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Cindy Q Yao
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Chad A Malloff
- Department of Integrative Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Lucia L Lam
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | - Honglei Xie
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Nicholas J Harding
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Denise Y F Mak
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Center for Addiction and Mental Health, Toronto, ON, Canada
| | - Kenneth C Chu
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Lauren C Chong
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Dorota H Sendorek
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Christine P'ng
- Informatics and Bio-Computing Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Colin C Collins
- Vancouver Prostate Centre and Department of Urological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jeremy A Squire
- Department of Pathology and Forensic Medicine, University of São Paulo at Ribeirão Preto, Ribeirão Preto, Brazil
| | - Igor Jurisica
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Colin Cooper
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK; Department of Biological Sciences and School of Medicine, University of East Anglia, Norwich, UK
| | - Rosalind Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK; Royal Marsden National Health Service Foundation Trust, London, UK
| | - Melania Pintilie
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, Bern University Hospital, Bern, Switzerland
| | - Elai Davicioni
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | - Wan L Lam
- Department of Integrative Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Michael Milosevic
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - David E Neal
- Uro-Oncology Research Group, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Department of Urology, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Theodorus van der Kwast
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Department of Pathology, Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Robert G Bristow
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
20
|
Abstract
Hyperpolarization using dissolution dynamic nuclear polarization has emerged as a versatile method to dramatically improve the MR signal of low-sensitivity nuclei. This technique facilitates the study of real-time metabolism in vitro and in vivo using (13)C-enriched substrates and has been applied to numerous models of human disease. In particular, several mechanisms underlying prostate cancer have been interrogated using hyperpolarized (13)C MR spectroscopy. This review highlights key metabolic shifts seen in prostate cancer, their study by hyperpolarized (13)C MR spectroscopy, and the development of new platforms for metabolic study.
Collapse
Affiliation(s)
- David M Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| |
Collapse
|
21
|
Chang AJ, Autio KA, Roach M, Scher HI. High-risk prostate cancer-classification and therapy. Nat Rev Clin Oncol 2014; 11:308-23. [PMID: 24840073 DOI: 10.1038/nrclinonc.2014.68] [Citation(s) in RCA: 296] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Approximately 15% of patients with prostate cancer are diagnosed with high-risk disease. However, the current definitions of high-risk prostate cancer include a heterogeneous group of patients with a range of prognoses. Some have the potential to progress to a lethal phenotype that can be fatal, while others can be cured with treatment of the primary tumour alone. The optimal management of this patient subgroup is evolving. A refined classification scheme is needed to enable the early and accurate identification of high-risk disease so that more-effective treatment paradigms can be developed. We discuss several principles established from clinical trials, and highlight other questions that remain unanswered. This Review critically evaluates the existing literature focused on defining the high-risk population, the management of patients with high-risk prostate cancer, and future directions to optimize care.
Collapse
Affiliation(s)
- Albert J Chang
- Department of Radiation Oncology, University of California, San Francisco, 1600 Divisadero Street, Suite H-1031, San Francisco, CA 94115, USA
| | - Karen A Autio
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mack Roach
- Department of Radiation Oncology, University of California, San Francisco, 1600 Divisadero Street, Suite H-1031, San Francisco, CA 94115, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
22
|
Fraser M, Berlin A, Bristow RG, van der Kwast T. Genomic, pathological, and clinical heterogeneity as drivers of personalized medicine in prostate cancer. Urol Oncol 2014; 33:85-94. [PMID: 24768356 DOI: 10.1016/j.urolonc.2013.10.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 12/23/2022]
Abstract
Prostate cancer (CaP) is the most commonly diagnosed malignancy in men in the Western world. In North America, more than 275,000 men are diagnosed annually, whereby approximately 1 in 6 men will be diagnosed with CaP in their lifetime, and 1 in 34 men will die from castration-resistant metastatic disease. Unfortunately, current clinical prognostic factors explain only a proportion of the observed variation in clinical outcome from patient to patient. Furthermore, overtreatment of indolent and low-risk cancers leads to inappropriate morbidity following radiotherapy or surgery. As such, better predictors of individualized prognosis and treatment response are urgently needed to triage patients to customized and intensified CaP treatment. Recent developments in next-generation sequencing have made it possible to identify prognostic and predictive signatures based on genomic profiles. We discuss the genetic basis of CaP progression from localized to systemic disease (e.g., point mutations, copy-number alterations, and structural variants) in relation with unique features of CaP biology, including intraprostatic and interprostatic heterogeneity, multifocality and multiclonality, TMPRSS2:ERG, and other ETS-family gene fusions. Finally, we focus on the use of genomic markers as prognostic factors for local failure and for systemic disease, as novel risk-stratification tools, in triaging patients to existing treatment options, and ultimately the potential of genomics for the identification of molecular targets for therapy of CaP.
Collapse
Affiliation(s)
- Michael Fraser
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada
| | - Alejandro Berlin
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Robert G Bristow
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Theodorus van der Kwast
- Department of Pathology and Laboratory Medicine, Toronto General Hospital (University Health Network), Toronto, Ontario, Canada.
| |
Collapse
|
23
|
Fontugne J, Lee D, Cantaloni C, Barbieri CE, Caffo O, Hanspeter E, Mazzoleni G, Dalla Palma P, Rubin MA, Fellin G, Mosquera JM, Barbareschi M, Demichelis F. Recurrent prostate cancer genomic alterations predict response to brachytherapy treatment. Cancer Epidemiol Biomarkers Prev 2014; 23:594-600. [PMID: 24515272 DOI: 10.1158/1055-9965.epi-13-1180] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the association of recurrent molecular alterations in prostate cancer, such as ERG rearrangements and phosphatase and tensin homolog gene (PTEN) deletions, with oncologic outcomes in patients with prostate cancer treated with brachytherapy. METHODS Ninety-two men underwent I-125 brachytherapy with a 145 Gy delivered dose between 2000 and 2008. Pretreatment prostate biopsies were analyzed by immunohistochemistry (IHC) and FISH for ERG rearrangement and overexpression, PTEN deletion, and expression loss. Univariable and multivariable Cox-regression analyses evaluated association of ERG and PTEN status with biochemical recurrence (BCR). RESULTS Within a median follow-up of 73 months, 11% of patients experienced BCR. Of 80 samples with both IHC and FISH performed for ERG, 46 (57.8%) demonstrated rearrangement by FISH and 45 (56.3%) by IHC. Of 77 samples with both IHC and FISH for PTEN, 14 (18.2%) had PTEN deletion by FISH and 22 (28.6%) by IHC. No significant associations were found between ERG, PTEN status, and clinicopathologic features. Patients with concurrent ERG rearrangement and PTEN deletion demonstrated significantly worse relapse-free survival rates compared with those with ERG or PTEN wild type (P < 0.01). In multivariable Cox regression analysis adjusted for the effects of standard clinicopathologic features, combined ERG rearranged and PTEN deletion was independently associated with BCR (HR = 2.6; P = 0.02). CONCLUSIONS Concurrent ERG rearrangement and PTEN loss was independently associated with time to BCR in patients undergoing brachytherapy. Future studies are needed to validate prostate cancer molecular subtyping for risk stratification. IMPACT Identifying patients in the ERG-rearranged/PTEN-deleted molecular subclass may improve treatment personalization.
Collapse
Affiliation(s)
- Jacqueline Fontugne
- Authors' Affiliations: Departments of Pathology and Laboratory Medicine and Urology; Institute for Precision Medicine; Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, New York; Centre for Integrative Biology, University of Trento; Departments of Medical Oncology, Pathology, and Radiotherapy and Medical Physics, Ospedale Santa Chiara, Trento; and Department of Surgical Pathology, Central Hospital, Bolzano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bristow RG, Berlin A, Dal Pra A. An arranged marriage for precision medicine: hypoxia and genomic assays in localized prostate cancer radiotherapy. Br J Radiol 2014; 87:20130753. [PMID: 24588670 DOI: 10.1259/bjr.20130753] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed malignancy in males in the Western world with one in six males diagnosed in their lifetime. Current clinical prognostication groupings use pathologic Gleason score, pre-treatment prostatic-specific antigen and Union for International Cancer Control-TNM staging to place patients with localized CaP into low-, intermediate- and high-risk categories. These categories represent an increasing risk of biochemical failure and CaP-specific mortality rates, they also reflect the need for increasing treatment intensity and justification for increased side effects. In this article, we point out that 30-50% of patients will still fail image-guided radiotherapy or surgery despite the judicious use of clinical risk categories owing to interpatient heterogeneity in treatment response. To improve treatment individualization, better predictors of prognosis and radiotherapy treatment response are needed to triage patients to bespoke and intensified CaP treatment protocols. These should include the use of pre-treatment genomic tests based on DNA or RNA indices and/or assays that reflect cancer metabolism, such as hypoxia assays, to define patient-specific CaP progression and aggression. More importantly, it is argued that these novel prognostic assays could be even more useful if combined together to drive forward precision cancer medicine for localized CaP.
Collapse
Affiliation(s)
- R G Bristow
- Princess Margaret Cancer Center (University Health Network), Toronto, ON, Canada
| | | | | |
Collapse
|
25
|
Dal Pra A, Lalonde E, Sykes J, Warde F, Ishkanian A, Meng A, Maloff C, Srigley J, Joshua AM, Petrovics G, van der Kwast T, Evans A, Milosevic M, Saad F, Collins C, Squire J, Lam W, Bismar TA, Boutros PC, Bristow RG. TMPRSS2-ERG Status Is Not Prognostic Following Prostate Cancer Radiotherapy: Implications for Fusion Status and DSB Repair. Clin Cancer Res 2013; 19:5202-9. [DOI: 10.1158/1078-0432.ccr-13-1049] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
McKee CM, Xu D, Cao Y, Kabraji S, Allen D, Kersemans V, Beech J, Smart S, Hamdy F, Ishkanian A, Sykes J, Pintile M, Milosevic M, van der Kwast T, Zafarana G, Ramnarine VR, Jurisica I, Mallof C, Lam W, Bristow RG, Muschel RJ. Protease nexin 1 inhibits hedgehog signaling in prostate adenocarcinoma. J Clin Invest 2012; 122:4025-36. [PMID: 23041623 DOI: 10.1172/jci59348] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 08/14/2012] [Indexed: 01/08/2023] Open
Abstract
Prostate adenocarcinoma (CaP) patients are classified into low-, intermediate-, and high-risk groups that reflect relative survival categories. While there are accepted treatment regimens for low- and high-risk patients, intermediate-risk patients pose a clinical dilemma, as treatment outcomes are highly variable for these individuals. A better understanding of the factors that regulate the progression of CaP is required to delineate risk. For example, aberrant activation of the Hedgehog (Hh) pathway is implicated in CaP progression. Here, we identify the serine protease inhibitor protease nexin 1 (PN1) as a negative regulator of Hh signaling in prostate. Using human CaP cell lines and a mouse xenograft model of CaP, we demonstrate that PN1 regulates Hh signaling by decreasing protein levels of the Hh ligand Sonic (SHH) and its downstream effectors. Furthermore, we show that SHH expression enhanced tumor growth while overexpression of PN1 inhibited tumor growth and angiogenesis in mice. Finally, using comparative genome hybridization, we found that genetic alterations in Hh pathway genes correlated with worse clinical outcomes in intermediate-risk CaP patients, indicating the importance of this pathway in CaP.
Collapse
Affiliation(s)
- Chad M McKee
- Gray Institute of Radiation Oncology and Biology, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Locke JA, Zafarana G, Malloff CA, Lam WL, Sykes J, Pintilie M, Ramnarine VR, Meng A, Ahmed O, Jurisica I, Guns ET, van der Kwast T, Milosevic M, Bristow RG. Allelic loss of the loci containing the androgen synthesis gene, StAR, is prognostic for relapse in intermediate-risk prostate cancer. Prostate 2012; 72:1295-305. [PMID: 22213075 DOI: 10.1002/pros.22478] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 12/02/2011] [Indexed: 12/22/2022]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) and novel agents targeting the androgen synthesis axis (e.g., abiraterone acetate) are adjuvant therapies that are currently, or may in the future be, combined with radiotherapy to reduce the chance of disease relapse. Little is known about allelic loss or gain pertaining to genes associated with the androgen synthesis axis and whether this is prognostic in patients who receive localized radiotherapy. In this hypothesis generating study, we conducted an array comparative genomic hybridization (aCGH) analysis of 33 androgen synthesis genes to identify potential prognostic factors for radiotherapy outcome. METHODS aCGH analysis of tumor DNA prospectively derived from frozen needle biopsies of 126 men with intermediate-risk disease who underwent image-guided radiotherapy (IGRT) to a mean dose of 76.4 Gy was conducted. Statistical analyses were conducted for allelic loss or gain in genes as potential prognostic factors relative to prostate specific antigen, Gleason-score, and T-category. RESULTS We observed that allelic losses of loci containing the genes StAR and HSD17B2 were associated with increased genetic instability (as determined by percentage genome alteration). On multivariate analyses these loci were prognostic for biochemical disease-free relapse (StAR: HR = 2.84, 95% CI: 1.44-5.61, P = 0.00269; HSD17B2: HR = 1.97, 95% CI: 1.06-3.64, P = 0.031). The results were validated in a surgical cohort of 131 intermediate-risk patients. CONCLUSIONS Allelic losses of the loci containing StAR and HSD17B2 have significant prognostic value for intermediate-risk prostate cancer. With this hypothesis generating information future studies should test StAR and HSD17B2 losses as biomarkers of androgen response in combined modality protocols.
Collapse
Affiliation(s)
- Jennifer A Locke
- Department of Radiation Oncology, Medical Biophysics, Laboratory Medicine and Pathology, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Thoms JW, Dal Pra A, Anborgh PH, Christensen E, Fleshner N, Menard C, Chadwick K, Milosevic M, Catton C, Pintilie M, Chambers AF, Bristow RG. Plasma osteopontin as a biomarker of prostate cancer aggression: relationship to risk category and treatment response. Br J Cancer 2012; 107:840-6. [PMID: 22871886 PMCID: PMC3425969 DOI: 10.1038/bjc.2012.345] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND High plasma osteopontin (OPN) has been linked to tumour hypoxia, metastasis, and poor prognosis. This study aims to assess whether plasma osteopontin was a biomarker of increasing progression within prostate cancer (PCa) prognostic groups and whether it reflected treatment response to local and systemic therapies. METHODS Baseline OPN was determined in men with localised (n=199), locally recurrent (n=9) and castrate-resistant, metastatic PCa (CRPC-MET; n=37). Receiver-operating curves (ROC) were generated to describe the accuracy of OPN for distinguishing between localised risk groups or localised vs metastatic disease. We also measured OPN pre- and posttreatment, following radical prostatectomy, external beam radiotherapy (EBRT), androgen deprivation (AD) or taxane-based chemotherapy. RESULTS The CRPC-MET patients had increased baseline values (mean 219; 56-513 ng ml(-1); P<0.0001) compared with the localised, non-metastatic group (mean 72; 12-438 ng ml(-1)). The area under the ROC to differentiate localised vs metastatic disease was improved when OPN was added to prostate-specific antigen (PSA) (0.943-0.969). Osteopontin neither distinguished high-risk PCa from other localised PCa nor correlated with serum PSA at baseline. Osteopontin levels reduced in low-risk patients after radical prostatectomy (P=0.005) and in CRPC-MET patients after chemotherapy (P=0.027), but not after EBRT or AD. CONCLUSION Plasma OPN is as good as PSA at predicting treatment response in CRPC-MET patients after chemotherapy. Our data do not support the use of plasma OPN as a biomarker of increasing tumour burden within localised PCa.
Collapse
Affiliation(s)
- J W Thoms
- Departments of Radiation Oncology, Surgery and Biostatistics, University of Toronto and Ontario Cancer Institute/Princess Margaret Hospital (University Health Network), Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Vesprini D, Catton C, Jacks L, Lockwood G, Rosewall T, Bayley A, Chung P, Gospodarowicz M, Ménard C, Milosevic M, Nichol A, Skala M, Warde P, Bristow RG. Inverse Relationship Between Biochemical Outcome and Acute Toxicity After Image-Guided Radiotherapy for Prostate Cancer. Int J Radiat Oncol Biol Phys 2012; 83:608-16. [DOI: 10.1016/j.ijrobp.2011.07.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 05/24/2011] [Accepted: 07/11/2011] [Indexed: 02/07/2023]
|
30
|
Zafarana G, Ishkanian AS, Malloff CA, Locke JA, Sykes J, Thoms J, Lam WL, Squire JA, Yoshimoto M, Ramnarine VR, Meng A, Ahmed O, Jurisica I, Jurisca I, Milosevic M, Pintilie M, van der Kwast T, Bristow RG. Copy number alterations of c-MYC and PTEN are prognostic factors for relapse after prostate cancer radiotherapy. Cancer 2012; 118:4053-62. [PMID: 22281794 DOI: 10.1002/cncr.26729] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 10/24/2011] [Accepted: 11/01/2011] [Indexed: 12/12/2022]
Abstract
Despite the use of PSA, Gleason score, and T-category as prognosticators in intermediate-risk prostate cancer, 20-40% of patients will fail local therapy. In order to optimize treatment approaches for intermediate-risk patients, additional genetic prognosticators are needed. Previous reports using array comparative genomic hybridization (aCGH) in radical prostatectomy cohorts suggested a combination of allelic loss of the PTEN gene on 10q and allelic gain of the c-MYC gene on 8q were associated with metastatic disease. We tested whether copy number alterations (CNAs) in PTEN (allelic loss) and c-MYC (allelic gain) were associated with biochemical relapse following modern-era, image-guided radiotherapy (mean dose 76.4 Gy). We used aCGH analyses validated by fluorescence in-situ hybridization (FISH) of DNA was derived from frozen, pre-treatment biopsies in 126 intermediate-risk prostate cancer patients. Patients whose tumors had CNAs in both PTEN and c-MYC had significantly increased genetic instability (percent genome alteration; PGA) compared to tumors with normal PTEN and c-MYC status (p < 0.0001). We demonstrate that c-MYC gain alone, or combined c-MYC gain and PTEN loss, were increasingly prognostic for relapse on multivariable analyses (hazard ratios (HR) of 2.58/p = 0.005 and 3.21/p = 0.0004; respectively). Triaging patients by the use of CNAs within pre-treatment biopsies may allow for better use of systemic therapies to target sub-clinical metastases or locally recurrent disease and improve clinical outcomes.
Collapse
Affiliation(s)
- Gaetano Zafarana
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Locke JA, Zafarana G, Ishkanian AS, Milosevic M, Thoms J, Have CL, Malloff CA, Lam WL, Squire JA, Pintilie M, Sykes J, Ramnarine VR, Meng A, Ahmed O, Jurisica I, van der Kwast T, Bristow RG. NKX3.1 haploinsufficiency is prognostic for prostate cancer relapse following surgery or image-guided radiotherapy. Clin Cancer Res 2011; 18:308-16. [PMID: 22048240 DOI: 10.1158/1078-0432.ccr-11-2147] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Despite the use of prostate specific antigen (PSA), Gleason-score, and T-category as prognostic factors, up to 40% of patients with intermediate-risk prostate cancer will fail radical prostatectomy or precision image-guided radiotherapy (IGRT). Additional genetic prognosticators are needed to triage these patients toward intensified combination therapy with novel targeted therapeutics. We tested the role of the NKX3.1 gene as a determinant of treatment outcome given its reported roles in tumor initiating cell (TIC) renewal, the DNA damage response, and cooperation with c-MYC during prostate cancer progression. METHODS Using high-resolution array comparative genomic hybridization (aCGH), we profiled the copy number alterations in TIC genes using tumor DNA from frozen needle biopsies derived from 126 intermediate-risk patients who underwent IGRT. These data were correlated to biochemical relapse-free rate (bRFR) by the Kaplan-Meier method and Cox proportional hazards models. RESULTS A screen of the aCGH-IGRT data for TIC genes showed frequent copy number alterations for NKX3.1, PSCA, and c-MYC. NKX3.1 haploinsufficiency was associated with increased genomic instability independent of PSA, T-category, and Gleason-score. After adjusting for clinical factors in a multivariate model, NKX3.1 haploinsufficiency was associated with bRFR when tested alone (HR = 3.05, 95% CI: 1.46-6.39, P = 0.0030) or when combined with c-MYC gain (HR = 3.88, 95% CI: 1.78-8.49, P = 0.00067). A similar association was observed for patients following radical prostatectomy with a public aCGH database. NKX3.1 status was associated with positive biopsies post-IGRT and increased clonogen radioresistance in vitro. CONCLUSIONS Our results support the use of genomic predictors, such as NKX3.1 status, in needle biopsies for personalized approaches to prostate cancer management.
Collapse
Affiliation(s)
- Jennifer A Locke
- Department of Radiation Oncology, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Qian Y, Feng FY, Halverson S, Blas K, Sandler HM, Hamstra DA. The Percent of Positive Biopsy Cores Improves Prediction of Prostate Cancer–Specific Death in Patients Treated With Dose-Escalated Radiotherapy. Int J Radiat Oncol Biol Phys 2011; 81:e135-42. [DOI: 10.1016/j.ijrobp.2011.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 01/01/2011] [Accepted: 01/03/2011] [Indexed: 10/18/2022]
|
33
|
Thoms J, Bristow RG. DNA repair targeting and radiotherapy: a focus on the therapeutic ratio. Semin Radiat Oncol 2011; 20:217-22. [PMID: 20832013 DOI: 10.1016/j.semradonc.2010.06.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Radiotherapy (RT) results in the production of a variety of ionizing radiation-induced lesion in DNA. Specific pathways of DNA repair are required to repair the variety of lesions, which include DNA single-strand breaks (SSBs), DNA double-strand breaks (DSBs), DNA base alterations, and DNA-DNA or DNA-protein cross-links. Nonrepaired DNA damage can lead to normal and tumor cell death via apoptosis, mitotic catastrophe, autophagy, or terminal growth arrest senescence. Targeting the sensing and repair of DNA damage is an exciting concept. This must be combined with precision RT to limit the volume of irradiated normal tissue, including the use of image-guided radiotherapy (IGRT) and brachytherapy. The therapeutic ratio of combined targeting of DNA combined with RT could also be preserved using biological approaches and includes the following: (1) the documentation of relative defects in DNA damage sensing and repair in malignant cells; (2) the preferential use of certain DNA repair pathways (eg, base excision repair or homologous recombination) in malignant tissues compare with normal tissues; (3) the targeting of repair defects in chronically hypoxic cells; and (4) optimal scheduling of a DNA repair inhibitor in the neoadjuvant, concurrent, or adjuvant combined treatment settings. In this review, we discuss the general rationale and the optimal timing and duration of DNA repair inhibition during fractionated RT with the emphasis on preserving the therapeutic ratio of cancer treatment.
Collapse
Affiliation(s)
- John Thoms
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
34
|
Ishkanian AS, Zafarana G, Thoms J, Bristow RG. Array CGH as a potential predictor of radiocurability in intermediate risk prostate cancer. Acta Oncol 2010; 49:888-94. [PMID: 20590366 DOI: 10.3109/0284186x.2010.499371] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Prostate cancer is the most common male cancer and up to one fifth of diagnosed patients will die of their disease. Current prognostic variables including T-category (of the TNM staging), the absolute or kinetics of prostatic specific antigen (PSA) and the pathologic Gleason score (GS) are utilized to place men in low, intermediate and high-risk prostate cancer risk groupings. There is great heterogeneity within the non-indolent intermediate risk group with respect to clinical response. It is therefore imperative that further genetic and other prognostic factors be identified to better individualize treatment. Somatic alterations in prostate cancer. Herein, we review the potential for somatic alterations in tumor-associated genes (based on comparative genomic hybridization (CGH) in prostate cancers to be novel prognostic, and possibly predictive, factors for prostate cancer radiotherapy response. Intermediate risk prostate cancers show alterations in a number of genes thought to be involved in radiosensitivity, DNA repair, cell death and stem cell renewal. These include deletions at 21q (TMPRSS2: ERG), 13q (RB1), 10q (PTEN), 8p (NKX3.1), additions at 8q21 (containing c-Myc)) and haplo-insufficiency for p53, PARP1, ATM and DNA-PKcs. Conclusions. The use of high-resolution CGH for fine-mapping of deletions and amplifications in pre-radiotherapy prostate cancer biopsies is feasible. Genetic alterations may delineate localized prostate cancer from systemic disease and be used as a predictive factor in that patients would be individually triaged to local (surgery versus radiotherapy) and/or adjuvant (adjuvant androgen ablation or post-operative radiotherapy) therapies in a prospective fashion to improve outcome. The knowledge of abnormal DNA repair pathways within in a given patient could allow for the judicious use of targeted agents (PARP/ATM inhibitors) as personalized medicine.
Collapse
Affiliation(s)
- Adrian S Ishkanian
- Department of Radiation Oncology, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
35
|
Tabassum A, Bristow RG, Venkateswaran V. Ingestion of selenium and other antioxidants during prostate cancer radiotherapy: A good thing? Cancer Treat Rev 2010; 36:230-4. [PMID: 20079573 DOI: 10.1016/j.ctrv.2009.12.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 12/07/2009] [Accepted: 12/09/2009] [Indexed: 01/25/2023]
Affiliation(s)
- A Tabassum
- Molecular and Cellular Biology, Sunnybrook Health Science Centre, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5.
| | | | | |
Collapse
|
36
|
Bristow RG. IMRT and molecular biological approaches in radiotherapy for prostate cancer. Eur J Cancer 2010; 45 Suppl 1:429-30. [PMID: 19775659 DOI: 10.1016/s0959-8049(09)70077-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Robert G Bristow
- Princess Margaret Hospital (University Health Network), Toronto, ON, Canada
| |
Collapse
|
37
|
Ishkanian AS, Mallof CA, Ho J, Meng A, Albert M, Syed A, van der Kwast T, Milosevic M, Yoshimoto M, Squire JA, Lam WL, Bristow RG. High-resolution array CGH identifies novel regions of genomic alteration in intermediate-risk prostate cancer. Prostate 2009; 69:1091-100. [PMID: 19350549 DOI: 10.1002/pros.20959] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Approximately one-third of prostate cancer patients present with intermediate risk disease. Interestingly, while this risk group is clinically well defined, it demonstrates the most significant heterogeneity in PSA-based biochemical outcome. Further, the majority of candidate genes associated with prostate cancer progression have been identified using cell lines, xenograft models, and high-risk androgen-independent or metastatic patient samples. We used a global high-resolution array comparative genomic hybridization (CGH) assay to characterize copy number alterations (CNAs) in intermediate risk prostate cancer. Herein, we show this risk group contains a number of alterations previously associated with high-risk disease: (1) deletions at 21q22.2 (TMPRSS2:ERG), 16q22-24 (containing CDH1), 13q14.2 (RB1), 10q23.31 (PTEN), 8p21 (NKX3.1); and, (2) amplification at 8q21.3-24.3 (containing c-MYC). In addition, we identified six novel microdeletions at high frequency: 1q42.12-q42.3 (33.3%), 5q12.3-13.3 (21%), 20q13.32-13.33 (29.2%), 22q11.21 (25%), 22q12.1 (29.2%), and 22q13.31 (33.3%). Further, we show there is little concordance between CNAs from these clinical samples and those found in commonly used prostate cancer cell models. These unexpected findings suggest that the intermediate-risk category is a crucial cohort warranting further study to determine if a unique molecular fingerprint can predict aggressive versus indolent phenotypes.
Collapse
Affiliation(s)
- Adrian S Ishkanian
- Department of Radiation Oncology, University of Toronto and Radiation Medicine Program, Princess Margaret Hospital-University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Christensen E, Evans KR, Ménard C, Pintilie M, Bristow RG. Practical approaches to proteomic biomarkers within prostate cancer radiotherapy trials. Cancer Metastasis Rev 2008; 27:375-85. [PMID: 18427732 DOI: 10.1007/s10555-008-9139-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Proteomic biomarkers may be useful for monitoring therapeutic response and to triage cancer patients to best therapy. OBJECTIVES In this review, we highlight the importance of specimen acquisition, preparation and analysis in radiotherapy proteomic studies. We also discuss practical approaches for the design and execution of clinical proteomic studies using our recent experience based on specimens accrued during prostate cancer radiation therapy. DISCUSSION AND CONCLUSIONS Numerous proteomic methods are being employed, including high-throughput mass spectrometry and immunoassays, and using solid tissues, blood and urine for analysis. Given the potential complexity of cytokine and other protein responses, there is a need to assess proteomic signatures within serial samples as longitudinal studies during a course of fractionated radiotherapy (RT).
Collapse
Affiliation(s)
- Eva Christensen
- Radiation Medicine Program, Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada.
| | | | | | | | | |
Collapse
|
39
|
Supiot S, Shubbar S, Fleshner N, Warde P, Hersey K, Wallace K, Cole H, Sweet J, Tsihlias J, Jewett MA, Klotz L, Bristow RG. A phase I trial of pre-operative radiotherapy for prostate cancer: Clinical and translational studies. Radiother Oncol 2008; 88:53-60. [DOI: 10.1016/j.radonc.2008.03.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 02/26/2008] [Accepted: 03/24/2008] [Indexed: 12/19/2022]
|
40
|
Lee JT, Lehmann BD, Terrian DM, Chappell WH, Stivala F, Libra M, Martelli AM, Steelman LS, McCubrey JA. Targeting prostate cancer based on signal transduction and cell cycle pathways. Cell Cycle 2008; 7:1745-62. [PMID: 18594202 DOI: 10.4161/cc.7.12.6166] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer remains a leading cause of death in men despite increased capacity to diagnose at earlier stages. After prostate cancer has become hormone independent, which often occurs after hormonal ablation therapies, it is difficult to effectively treat. Prostate cancer may arise from mutations and dysregulation of various genes involved in regulation signal transduction (e.g., PTEN, Akt, etc.,) and the cell cycle (e.g., p53, p21(Cip1), p27(Kip1), Rb, etc.,). This review focuses on the aberrant interactions of signal transduction and cell cycle genes products and how they can contribute to prostate cancer and alter therapeutic effectiveness.
Collapse
Affiliation(s)
- John T Lee
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, North Carolina 27858, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lehmann BD, McCubrey JA, Terrian DM. Radiosensitization of prostate cancer by priming the wild-type p53-dependent cellular senescence pathway. Cancer Biol Ther 2007; 6:1165-70. [PMID: 18059157 DOI: 10.4161/cbt.6.8.4544] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A dramatic stage-migration in diagnosis of prostate cancer has led to earlier detection of clinically localized carcinoma and an increased use of radiation therapy. The p53 protein responds to irradiation-induced DNA damage by removing critically damaged cells from the proliferative pool. This review will focus on the dominant role that p53-dependent cellular senescence, rather than cell death, plays in determining the radiosensitivity of human prostate cancer cells in vitro. The finding that senescence is a primary mechanism of tumor regression indicates that p53 activators or downstream effectors may prove effective in radiosensitizing some carcinoma of the prostate.
Collapse
Affiliation(s)
- Brian D Lehmann
- Departments of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina, USA
| | | | | |
Collapse
|
42
|
Jereczek-Fossa BA, Orecchia R. Evidence-based radiation oncology: Definitive, adjuvant and salvage radiotherapy for non-metastatic prostate cancer. Radiother Oncol 2007; 84:197-215. [PMID: 17532494 DOI: 10.1016/j.radonc.2007.04.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Revised: 04/08/2007] [Accepted: 04/18/2007] [Indexed: 02/07/2023]
Abstract
The standard treatment options based on the risk category (stage, Gleason score, PSA) for localized prostate cancer include surgery, radiotherapy and watchful waiting. The literature does not provide clear-cut evidence for the superiority of surgery over radiotherapy, whereas both approaches differ in their side effects. The definitive external beam irradiation is frequently employed in stage T1b-T1c, T2 and T3 tumors. There is a pretty strong evidence that intermediate- and high-risk patients benefit from dose escalation. The latter requires reduction of the irradiated normal tissue (using 3-dimensional conformal approach, intensity modulated radiotherapy, image-guided radiotherapy, etc.). Recent data suggest that prostate cancer may benefit from hypofractionation due to relatively low alpha/beta ratio; these findings warrant confirmation though. The role of whole pelvis irradiation is still controversial. Numerous randomized trials demonstrated a clinical benefit in terms of biochemical control, local and distant control, and overall survival from the addition of androgen suppression to external beam radiotherapy in intermediate- and high-risk patients. These studies typically included locally advanced (T3-T4) and poor-prognosis (Gleason score >7 and/or PSA >20 ng/mL) tumors and employed neoadjuvant/concomitant/adjuvant androgen suppression rather than only adjuvant setting. The ongoing trials will hopefully further define the role of endocrine treatment in more favorable risk patients and in the setting of the dose escalated radiotherapy. Brachytherapy (BRT) with permanent implants may be offered to low-risk patients (cT1-T2a, Gleason score <7, or 3+4, PSA <or=10 ng/mL), with prostate volume of <or=50 ml, no previous transurethral prostate resection and a good urinary function. Some recent data suggest a benefit from combining external beam irradiation and BRT for intermediate-risk patients. EBRT after radical prostatectomy improves disease-free survival and biochemical and local control rates in patients with positive surgical margins or pT3 tumors. Salvage radiotherapy may be considered at the time of biochemical failure in previously non-irradiated patients.
Collapse
|
43
|
Chan N, Milosevic M, Bristow RG. Tumor hypoxia, DNA repair and prostate cancer progression: new targets and new therapies. Future Oncol 2007; 3:329-41. [PMID: 17547528 DOI: 10.2217/14796694.3.3.329] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Increasingly, the tumor microenvironment and hypoxia are being studied as potential prognostic factors in prostate cancer given their effects on the hypoxia inducible factor-1α and vascular endothelial growth factor signaling pathways. Based on immunohistochemical studies using hypoxic cell markers and direct oxygen-electrode measurements, clinically relevant levels of hypoxia are detected in 30–90% of prostate cancers. Exciting new data suggest that hypoxia can alter cell-cycle checkpoints and DNA repair within the prostate epithelium, thereby driving genetic instability and tumor aggression. Novel therapies designed to target the hypoxic response and resulting defective DNA repair may therefore be effective as chemoprevention agents or as adjuncts to surgery, radiotherapy and chemotherapy to improve clinical outcome.
Collapse
Affiliation(s)
- Norman Chan
- Departments of Medical Biophysics & Radiation Oncology, University of Toronto, Princess Margaret Hospital (University Health Network), Toronto, Ontario, Canada.
| | | | | |
Collapse
|
44
|
Bristow RG, Ozcelik H, Jalali F, Chan N, Vesprini D. Homologous recombination and prostate cancer: a model for novel DNA repair targets and therapies. Radiother Oncol 2007; 83:220-30. [PMID: 17531338 DOI: 10.1016/j.radonc.2007.04.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2007] [Accepted: 04/17/2007] [Indexed: 01/30/2023]
Abstract
Using elegant targeting techniques such as IMRT, radiation oncology has improved the therapeutic ratio of prostate cancer radiotherapy through increased physical precision (e.g. increased local control through dose-escalation without increased normal tissue toxicity). The therapeutic ratio might be further improved by the addition of "biologic precision and escalation" pertaining to the use of molecular inhibitors of DNA damage sensing and repair. Indeed, proteins involved in the ATM-p53 damage signaling axis and the homologous (HR) and non-homologous end-joining (NHEJ) pathways of DNA double-strand break (DNA-dsb) rejoining pathways may be attractive candidates to elucidate cancer risk, prognosis, prediction of response and to develop sensitizers towards oxic and hypoxic prostate tumor cells. This review highlights DNA-dsb in prostate cancer research in terms of novel molecular inhibitors, the role of the microenvironment in DNA-dsb repair and potential DNA-dsb biomarkers for clinical trials.
Collapse
Affiliation(s)
- Robert G Bristow
- Department of Medical Biophysics, University of Toronto and Princess Margaret Hospital (University Health Network), Toronto, Canada.
| | | | | | | | | |
Collapse
|
45
|
Pinthus JH, Bryskin I, Trachtenberg J, Lu JP, Singh G, Fridman E, Wilson BC. Androgen induces adaptation to oxidative stress in prostate cancer: implications for treatment with radiation therapy. Neoplasia 2007; 9:68-80. [PMID: 17325745 PMCID: PMC1803036 DOI: 10.1593/neo.06739] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Revised: 12/21/2006] [Accepted: 12/27/2006] [Indexed: 01/14/2023] Open
Abstract
Radiation therapy is a standard treatment for prostate cancer (PC). The postulated mechanism of action for radiation therapy is the generation of reactive oxygen species (ROS). Adjuvant androgen deprivation (AD) therapy has been shown to confer a survival advantage over radiation alone in high-risk localized PC. However, the mechanism of this interaction is unclear. We hypothesize that androgens modify the radioresponsiveness of PC through the regulation of cellular oxidative homeostasis. Using androgen receptor (AR)(+) 22rv1 and AR(-) PC3 human PC cell lines, we demonstrated that testosterone increased basal reactive oxygen species (bROS) levels, resulting in dose-dependent activation of phospho-p38 and pAKT, and increased expression of clusterin, catalase, and manganese superoxide dismutase. Similar data were obtained in three human PC xenografts; WISH-PC14, WISH-PC23, and CWR22, growing in testosterone-supplemented or castrated SCID mice. These effects were reversible through AD or through incubation with a reducing agent. Moreover, testosterone increased the activity of catalase, superoxide dismutases, and glutathione reductase. Consequently, AD significantly facilitated the response of AR(+) cells to oxidative stress challenge. Thus, testosterone induces a preset cellular adaptation to radiation through the generation of elevated bROS, which is modified by AD. These findings provide a rational for combined hormonal and radiation therapy for localized PC.
Collapse
Affiliation(s)
- Jehonathan H Pinthus
- The Prostate Cancer Center, University Health Network, Toronto, Ontario, Canada.
| | | | | | | | | | | | | |
Collapse
|