51
|
Nafissi NN, Kosiorek HE, Butterfield RJ, Moore C, Ho T, Singh P, Bryce AH. Evolving Natural History of Metastatic Prostate Cancer. Cureus 2020; 12:e11484. [PMID: 33329980 PMCID: PMC7735525 DOI: 10.7759/cureus.11484] [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] [Indexed: 12/03/2022] Open
Abstract
Introduction The systemic therapies available to patients with metastatic prostate cancer (mPC) have improved dramatically over the past decade. Anecdotal experience suggests that the increased available lines of therapy have changed the profile of mPC to include a higher prevalence of visceral metastases. Materials and Methods A retrospective review of 472 patients with prostate cancer who died in 2009 and in 2016 was performed. Patients with metastatic disease who had imaging within six months of death were included. A total of 164 patients were eligible for analysis. Results Overall rates of visceral and distant metastases, including the lung, liver, adrenal, brain, renal, spleen, and thyroid, were higher in patients who died in 2016 as compared to those who died in 2009 (40.0% and 26.1%, respectively, p-value = 0.07). Forty-four percent of patients who died in 2016 used five or more lines of systemic treatments compared to 26.1% of patients in 2009. Conclusion The emergence of new systemic therapies for mPC is changing the natural history of the disease. Visceral metastases are being seen with increasing frequency than in the past. This observation is important for clinicians who are monitoring patients with prostate cancer to maintain a high suspicion for visceral disease.
Collapse
|
52
|
Opening a Scan of Worms. Eur Urol Oncol 2020; 3:725-727. [PMID: 33139238 DOI: 10.1016/j.euo.2020.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 10/16/2020] [Indexed: 11/22/2022]
|
53
|
Tunariu N, Blackledge M, Messiou C, Petralia G, Padhani A, Curcean S, Curcean A, Koh DM. What's New for Clinical Whole-body MRI (WB-MRI) in the 21st Century. Br J Radiol 2020; 93:20200562. [PMID: 32822545 PMCID: PMC8519652 DOI: 10.1259/bjr.20200562] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Whole-body MRI (WB-MRI) has evolved since its first introduction in the 1970s as an imaging technique to detect and survey disease across multiple sites and organ systems in the body. The development of diffusion-weighted MRI (DWI) has added a new dimension to the implementation of WB-MRI on modern scanners, offering excellent lesion-to-background contrast, while achieving acceptable spatial resolution to detect focal lesions 5 to 10 mm in size. MRI hardware and software advances have reduced acquisition times, with studies taking 40-50 min to complete.The rising awareness of medical radiation exposure coupled with the advantages of MRI has resulted in increased utilization of WB-MRI in oncology, paediatrics, rheumatological and musculoskeletal conditions and more recently in population screening. There is recognition that WB-MRI can be used to track disease evolution and monitor response heterogeneity in patients with cancer. There are also opportunities to combine WB-MRI with molecular imaging on PET-MRI systems to harness the strengths of hybrid imaging. The advent of artificial intelligence and machine learning will shorten image acquisition times and image analyses, making the technique more competitive against other imaging technologies.
Collapse
Affiliation(s)
| | - Matthew Blackledge
- Department of Radiotherapy, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London, UK
| | - Christina Messiou
- Department of Radiology, Royal Marsden Hospital, Downs Road, Sutton, London, UK
| | - Giuseppe Petralia
- Department of Radiology, European Institute of Oncology, Via Ripamonti, 435 - 20141 Milan, Italy
| | - Anwar Padhani
- Mount Vernon Hospital, The Paul Strickland Scanner Centre, Rickmansworth Road, Northwood, Middlesex, UK
| | - Sebastian Curcean
- Department of Radiology, Royal Marsden Hospital, Downs Road, Sutton, London, UK
| | | | - Dow-Mu Koh
- Drug Development Unit, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London, UK
| |
Collapse
|
54
|
Whole-body magnetic resonance imaging (WB-MRI) reporting with the METastasis Reporting and Data System for Prostate Cancer (MET-RADS-P): inter-observer agreement between readers of different expertise levels. Cancer Imaging 2020; 20:77. [PMID: 33109268 PMCID: PMC7590732 DOI: 10.1186/s40644-020-00350-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/24/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The METastasis Reporting and Data System for Prostate Cancer (MET-RADS-P) guidelines are designed to enable reproducible assessment in detecting and quantifying metastatic disease response using whole-body magnetic resonance imaging (WB-MRI) in patients with advanced prostate cancer (APC). The purpose of our study was to evaluate the inter-observer agreement of WB-MRI examination reports produced by readers of different expertise when using the MET-RADS-P guidelines. METHODS Fifty consecutive paired WB-MRI examinations, performed from December 2016 to February 2018 on 31 patients, were retrospectively examined to compare reports by a Senior Radiologist (9 years of experience in WB-MRI) and Resident Radiologist (after a 6-months training) using MET-RADS-P guidelines, for detection and for primary/dominant and secondary response assessment categories (RAC) scores assigned to metastatic disease in 14 body regions. Inter-observer agreement regarding RAC score was evaluated for each region by using weighted-Cohen's Kappa statistics (K). RESULTS The number of metastatic regions reported by the Senior Radiologist (249) and Resident Radiologist (251) was comparable. For the primary/dominant RAC pattern, the agreement between readers was excellent for the metastatic findings in cervical, dorsal, and lumbosacral spine, pelvis, limbs, lungs and other sites (K:0.81-1.0), substantial for thorax, retroperitoneal nodes, other nodes and liver (K:0.61-0.80), moderate for pelvic nodes (K:0.56), fair for primary soft tissue and not assessable for skull due to the absence of findings. For the secondary RAC pattern, agreement between readers was excellent for the metastatic findings in cervical spine (K:0.93) and retroperitoneal nodes (K:0.89), substantial for those in dorsal spine, pelvis, thorax, limbs and pelvic nodes (K:0.61-0.80), and moderate for lumbosacral spine (K:0.44). CONCLUSIONS We found inter-observer agreement between two readers of different expertise levels to be excellent in bone, but mixed in other body regions. Considering the importance of bone metastases in patients with APC, our results favor the use of MET-RADS-P in response to the growing clinical need for monitoring of metastasis in these patients.
Collapse
|
55
|
Bryce AH, Chen YH, Liu G, Carducci MA, Jarrard DM, Garcia JA, Dreicer R, Hussain M, Eisenberger MA, Plimack ER, Vogelzang NJ, DiPaola RS, Harshman L, Sweeney CJ. Patterns of Cancer Progression of Metastatic Hormone-sensitive Prostate Cancer in the ECOG3805 CHAARTED Trial. Eur Urol Oncol 2020; 3:717-724. [PMID: 32807727 DOI: 10.1016/j.euo.2020.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/11/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND ECOG3805 is a randomized trial of testosterone suppression with or without docetaxel for metastatic hormone-sensitive prostate cancer (mHSPC). Deeper prostate-specific antigen (PSA) suppression is prognostic for outcome. However, the concordance of PSA rise and radiographic progression has not been examined previously in mHSPC, whereas this has been reported in metastatic castration-resistant prostate cancer. OBJECTIVE To determine the patterns of progression by PSA and radiographic parameters in patients in ECOG3805. DESIGN, SETTING, AND PARTICIPANTS We conducted a retrospective analysis of all patients in ECOG3805. Patients were classified according to the PSA level at progression (whether PSA level was below 2.0 ng/mL or not) and the type of progression event in the study (either PSA progression as defined by the study with or without clinical progression, or clinical progression alone). Baseline demographics, clinical outcomes, and patterns of progression were compared between the groups. RESULTS AND LIMITATIONS One in eight patients had clinical progression below a PSA level of 2 ng/mL, and approximately 25% developed clinical progression in the absence of confirmed PSA progression. Overall survival from randomization was shorter in patients with clinical progression without confirmed PSA progression than in patients with PSA progression alone as the first progression. Patient demographics at study entry were not predictive of the pattern of progression. Study limitations include its retrospective and post hoc nature. CONCLUSIONS Clinical progression prior to PSA rise or at low PSA levels is a relatively frequent phenomenon in mHSPC and is associated with poorer overall survival. Further biological and clinical studies of these patients are warranted. PATIENT SUMMARY Reliance on prostate-specific antigen (PSA) alone is an inadequate strategy to monitor patients undergoing treatment for metastatic hormone-sensitive prostate cancer. Prostate cancer can get worse on scans even with low PSA and/or no or small changes in PSA. Imaging should be added to PSA testing to monitor patients with metastatic prostate cancer.
Collapse
Affiliation(s)
- Alan H Bryce
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA.
| | - Yu Hui Chen
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Glenn Liu
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | | | - David M Jarrard
- Department of Urology, University of Wisconsin, Madison, WI, USA
| | - Jorge A Garcia
- Department of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | | | - Maha Hussain
- Division of Hematology Oncology, Northwestern University, Chicago, IL, USA
| | | | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | - Lauren Harshman
- Department of Medicine, Dana-Farber Cancer Institute, Boston, MA, USA
| | | |
Collapse
|
56
|
Liquid Biopsy by Next-Generation Sequencing: a Multimodality Test for Management of Cancer. Curr Hematol Malig Rep 2020; 14:358-367. [PMID: 31346903 DOI: 10.1007/s11899-019-00532-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW While liquid biopsy is still relatively a new concept, the advent of next-generation sequencing (NGS) technologies has recently generated a revolution in the field and will be the focus of this review. RECENT FINDINGS Circulating tumor DNA (ctDNA) derives from tumor cells and provides information about the genetic alterations of tumors. However, ctDNA concentration in plasma can be below the level of detection by conventional methods; therefore, screening for actionable genetic information is challenging. Clinical trials exploring targeted and untargeted sequencing to improve the outcomes of ctDNA detection are showing promising results, having reached a limit of detection as low as 0.001% of ctDNA in a background of normal circulating DNA. Most of the challenges related to the sensitivity of detection of ctDNA have been defeated by dint of NGS-based approaches. Despite all the efforts, these methods are still expensive, time-consuming, and require advanced skills for appropriate interpretation. Nevertheless, the technology is rapidly improving, and the expectations for the implementation of liquid biopsy into the clinical practice in the near future are high.
Collapse
|
57
|
Tanaka T, Yang M, Froemming AT, Bryce AH, Inai R, Kanazawa S, Kawashima A. Current Imaging Techniques for and Imaging Spectrum of Prostate Cancer Recurrence and Metastasis: A Pictorial Review. Radiographics 2020; 40:709-726. [PMID: 32196428 DOI: 10.1148/rg.2020190121] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Relapsing level of prostate-specific antigen (PSA) after initial curative-intent local therapy for organ-confined prostate cancer is often the first sign of recurrence. However, PSA level recurrence does not enable accurate differentiation of locally recurrent tumor from metastatic disease or a combination of both. Metastatic prostate cancer most frequently involves bones and lymph nodes, followed by other organs such as the liver, lung, pleura, adrenal gland, ureter, peritoneum, penis, testis, and meninges. Conventional imaging including CT and bone scintigraphy has long been the standard of care but has limited sensitivity in depicting early local recurrence or metastatic disease. Multiparametric MRI has been shown to be more sensitive in detecting locally recurrent tumor in the prostatectomy bed as well as in situ recurrence in a prostate gland that has been treated with radiation therapy or thermal ablation. In addition, lesions detected with multiparametric MRI may be amenable to targeted biopsy for definitive diagnosis of recurrence. PET/CT or PET/MRI using the U.S. Food and Drug Administration (FDA)-approved tracers carbon 11 choline or fluorine 18 fluciclovine has demonstrated markedly increased sensitivity and specificity for diagnosis of early metastatic disease such as small-volume lymph node metastasis, as have a range of investigational gallium 68 prostate-specific membrane antigen (PSMA) radioactive PET tracers. With recent advances in imaging modalities and techniques, more accurate early detection, localization, and characterization of recurrent prostate cancer have become possible. The authors present a contemporary review of the strengths and limitations of conventional and advanced imaging modalities in evaluation of patients with recurrent prostate cancer and a systematic review of the clinical and imaging features of locally recurrent and metastatic disease.©RSNA, 2020See discussion on this article by Barwick and Castellucci.
Collapse
Affiliation(s)
- Takashi Tanaka
- From the Department of Radiology (T.T., M.Y., A.K.) and Department of Hematology and Medical Oncology (A.H.B.), Mayo Clinic, Scottsdale, Ariz; Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan (T.T., R.I., S.K.); and Department of Radiology, Mayo Clinic, Rochester, Minn (A.T.F.)
| | - Ming Yang
- From the Department of Radiology (T.T., M.Y., A.K.) and Department of Hematology and Medical Oncology (A.H.B.), Mayo Clinic, Scottsdale, Ariz; Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan (T.T., R.I., S.K.); and Department of Radiology, Mayo Clinic, Rochester, Minn (A.T.F.)
| | - Adam T Froemming
- From the Department of Radiology (T.T., M.Y., A.K.) and Department of Hematology and Medical Oncology (A.H.B.), Mayo Clinic, Scottsdale, Ariz; Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan (T.T., R.I., S.K.); and Department of Radiology, Mayo Clinic, Rochester, Minn (A.T.F.)
| | - Alan H Bryce
- From the Department of Radiology (T.T., M.Y., A.K.) and Department of Hematology and Medical Oncology (A.H.B.), Mayo Clinic, Scottsdale, Ariz; Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan (T.T., R.I., S.K.); and Department of Radiology, Mayo Clinic, Rochester, Minn (A.T.F.)
| | - Ryota Inai
- From the Department of Radiology (T.T., M.Y., A.K.) and Department of Hematology and Medical Oncology (A.H.B.), Mayo Clinic, Scottsdale, Ariz; Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan (T.T., R.I., S.K.); and Department of Radiology, Mayo Clinic, Rochester, Minn (A.T.F.)
| | - Susumu Kanazawa
- From the Department of Radiology (T.T., M.Y., A.K.) and Department of Hematology and Medical Oncology (A.H.B.), Mayo Clinic, Scottsdale, Ariz; Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan (T.T., R.I., S.K.); and Department of Radiology, Mayo Clinic, Rochester, Minn (A.T.F.)
| | - Akira Kawashima
- From the Department of Radiology (T.T., M.Y., A.K.) and Department of Hematology and Medical Oncology (A.H.B.), Mayo Clinic, Scottsdale, Ariz; Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama 700-8558, Japan (T.T., R.I., S.K.); and Department of Radiology, Mayo Clinic, Rochester, Minn (A.T.F.)
| |
Collapse
|
58
|
Gillessen S, Attard G, Beer TM, Beltran H, Bjartell A, Bossi A, Briganti A, Bristow RG, Chi KN, Clarke N, Davis ID, de Bono J, Drake CG, Duran I, Eeles R, Efstathiou E, Evans CP, Fanti S, Feng FY, Fizazi K, Frydenberg M, Gleave M, Halabi S, Heidenreich A, Heinrich D, Higano CTS, Hofman MS, Hussain M, James N, Kanesvaran R, Kantoff P, Khauli RB, Leibowitz R, Logothetis C, Maluf F, Millman R, Morgans AK, Morris MJ, Mottet N, Mrabti H, Murphy DG, Murthy V, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Parker C, Poon DMC, Pritchard CC, Reiter RE, Roach M, Rubin M, Ryan CJ, Saad F, Sade JP, Sartor O, Scher HI, Shore N, Small E, Smith M, Soule H, Sternberg CN, Steuber T, Suzuki H, Sweeney C, Sydes MR, Taplin ME, Tombal B, Türkeri L, van Oort I, Zapatero A, Omlin A. Management of Patients with Advanced Prostate Cancer: Report of the Advanced Prostate Cancer Consensus Conference 2019. Eur Urol 2020; 77:508-547. [PMID: 32001144 DOI: 10.1016/j.eururo.2020.01.012] [Citation(s) in RCA: 267] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Innovations in treatments, imaging, and molecular characterisation in advanced prostate cancer have improved outcomes, but there are still many aspects of management that lack high-level evidence to inform clinical practice. The Advanced Prostate Cancer Consensus Conference (APCCC) 2019 addressed some of these topics to supplement guidelines that are based on level 1 evidence. OBJECTIVE To present the results from the APCCC 2019. DESIGN, SETTING, AND PARTICIPANTS Similar to prior conferences, experts identified 10 important areas of controversy regarding the management of advanced prostate cancer: locally advanced disease, biochemical recurrence after local therapy, treating the primary tumour in the metastatic setting, metastatic hormone-sensitive/naïve prostate cancer, nonmetastatic castration-resistant prostate cancer, metastatic castration-resistant prostate cancer, bone health and bone metastases, molecular characterisation of tissue and blood, inter- and intrapatient heterogeneity, and adverse effects of hormonal therapy and their management. A panel of 72 international prostate cancer experts developed the programme and the consensus questions. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The panel voted publicly but anonymously on 123 predefined questions, which were developed by both voting and nonvoting panel members prior to the conference following a modified Delphi process. RESULTS AND LIMITATIONS Panellists voted based on their opinions rather than a standard literature review or formal meta-analysis. The answer options for the consensus questions had varying degrees of support by the panel, as reflected in this article and the detailed voting results reported in the Supplementary material. CONCLUSIONS These voting results from a panel of prostate cancer experts can help clinicians and patients navigate controversial areas of advanced prostate management for which high-level evidence is sparse. However, diagnostic and treatment decisions should always be individualised based on patient-specific factors, such as disease extent and location, prior lines of therapy, comorbidities, and treatment preferences, together with current and emerging clinical evidence and logistic and economic constraints. Clinical trial enrolment for men with advanced prostate cancer should be strongly encouraged. Importantly, APCCC 2019 once again identified important questions that merit assessment in specifically designed trials. PATIENT SUMMARY The Advanced Prostate Cancer Consensus Conference provides a forum to discuss and debate current diagnostic and treatment options for patients with advanced prostate cancer. The conference, which has been held three times since 2015, aims to share the knowledge of world experts in prostate cancer management with health care providers worldwide. At the end of the conference, an expert panel discusses and votes on predefined consensus questions that target the most clinically relevant areas of advanced prostate cancer treatment. The results of the voting provide a practical guide to help clinicians discuss therapeutic options with patients as part of shared and multidisciplinary decision making.
Collapse
Affiliation(s)
- Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Universita della Svizzera Italiana, Lugano, Switzerland; Cantonal Hospital, St. Gallen, Switzerland; University of Bern, Bern, Switzerland; Division of Cancer Science, University of Manchester, Manchester, UK.
| | | | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Himisha Beltran
- Dana-Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anders Bjartell
- Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Alberto Bossi
- Genito Urinary Oncology, Prostate Brachytherapy Unit, Goustave Roussy, Paris, France
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, URI, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Rob G Bristow
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Christie NHS Trust, Manchester, UK; CRUK Manchester Institute and Cancer Centre, Manchester, UK
| | - Kim N Chi
- BC Cancer, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Noel Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - Ian D Davis
- Monash University and Eastern Health, Victoria, Australia
| | - Johann de Bono
- The Institute of Cancer Research/Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Charles G Drake
- Division of Haematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | - Ignacio Duran
- Department of Medical Oncology, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Cantabria, Spain
| | - Ros Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | | | | | | | - Felix Y Feng
- University of California San Francisco, San Francisco, CA, USA
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Sud, Villejuif, France
| | - Mark Frydenberg
- Department of Surgery, Monash University, Melbourne, Australia; Prostate Cancer Research Program, Monash University, Melbourne, Australia; Department Anatomy & Developmental Biology, Faculty of Nursing, Medicine & Health Sciences, Monash University, Melbourne, Australia
| | - Martin Gleave
- Urological Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Axel Heidenreich
- Department of Urology, Uro-Oncology, Robot-Assisted and Reconstructive Urology, University of Cologne, Cologne, Germany; Department of Urology, Medical University, Vienna, Austria
| | - Daniel Heinrich
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Celestia Tia S Higano
- University of Washington, Seattle, WA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michael S Hofman
- Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | | | | | - Philip Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Raja B Khauli
- Department of Urology, American University of Beirut Medical Center, Beirut, Lebanon; Naef K. Basile Cancer Institute (NKBCI), American University of Beirut Medical Center, Beirut, Lebanon
| | - Raya Leibowitz
- Oncology institute, Shamir Medical Center and Faculty of medicine, Tel-Aviv University, Israel
| | - Chris Logothetis
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Centre, Houston, TX, USA; Department of Clinical Therapeutics, David H. Koch Centre, University of Athens Alexandra Hospital, Athens, Greece
| | - Fernando Maluf
- Beneficiência Portuguesa de São Paulo, São Paulo, SP, Brazil; Departamento de Oncologia, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Alicia K Morgans
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | | | | | - Hind Mrabti
- National Institute of Oncology, University hospital, Rabat, Morocco
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | | | - William K Oh
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| | - Piet Ost
- Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Joe M O'Sullivan
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland, UK; Radiotherapy Department, Cancer Centre, Belfast City Hospital, Belfast, Northern Ireland, UK
| | - Anwar R Padhani
- Mount Vernon Cancer Centre and Institute of Cancer Research, London, UK
| | - Chris Parker
- Royal Marsden Hospital and Institute of Cancer Research, Sutton, UK
| | - Darren M C Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | | | - Mack Roach
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Mark Rubin
- Bern Center for Precision Medicine, Bern, Switzerland; Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Charles J Ryan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Fred Saad
- Centre Hospitalier de Université de Montréal, Montreal, Canada
| | | | | | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Matthew Smith
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | - Cora N Sternberg
- Division of Hematology and Oncology, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Thomas Steuber
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Christopher Sweeney
- Dana-Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Levent Türkeri
- Department of Urology, M.A. Aydınlar Acıbadem University, Altunizade Hospital, Istanbul, Turkey
| | - Inge van Oort
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Almudena Zapatero
- Department of Radiation Oncology, University Hospital La Princesa, Health Research Institute, Madrid, Spain
| | - Aurelius Omlin
- University of Bern, Bern, Switzerland; Department of Medical Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
59
|
Clinical progression is associated with poor prognosis whatever the treatment line in metastatic castration resistant prostate cancer: The CATS international database. Eur J Cancer 2020; 125:153-163. [DOI: 10.1016/j.ejca.2019.10.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 10/30/2019] [Indexed: 11/17/2022]
|
60
|
Lapini A, Caffo O, Pappagallo G, Iacovelli R, D’Angelillo RM, Vavassori V, Ceccarelli R, Bracarda S, Jereczek-Fossa BA, Da Pozzo L, Conti GN. Monitoring Patients with Metastatic Hormone-Sensitive and Metastatic Castration-Resistant Prostate Cancer: A Multidisciplinary Consensus Document. Cancers (Basel) 2019; 11:cancers11121908. [PMID: 31805687 PMCID: PMC6966424 DOI: 10.3390/cancers11121908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 11/24/2022] Open
Abstract
Background: The availability of a number of agents that are efficacious in patients with metastatic prostate cancer (mPC) has led to them being used sequentially, and this has prolonged patient survival. However, in order to maximize their efficacy, clinicians need to be able to obtain a reliable picture of disease evolution by means of monitoring procedures. Methods: As the intensive monitoring protocols used in pivotal trials cannot be adopted in everyday clinical practice and there is no agreement among the available guidelines, a multidisciplinary panel of Italian experts met to develop recommendations for monitoring mPC patients using a modified Delphi method. Results: The consensus project considered methods of clinically, radiographically, and biochemically monitoring patients with metastatic hormone-sensitive and metastatic castration-resistant prostate cancer undergoing chemotherapy and/or hormonal treatment. The panelists also considered the methods and timing of monitoring castration levels, bone health, and the metabolic syndrome during androgen deprivation therapy. Conclusions: The recommendations, which were drawn up by experts following a formal and validated consensus procedure, will help clinicians face the everyday challenges of monitoring metastatic prostate cancer patients.
Collapse
Affiliation(s)
- Alberto Lapini
- Department of Urology, University of Florence, University Hospital, 50134 Florence, Italy;
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, 38112 Trento, Italy
- Correspondence: ; Tel.: +39-0461-902478
| | | | - Roberto Iacovelli
- Department of Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | | | - Vittorio Vavassori
- Department of Radiation Oncology, Humanitas Gavazzeni, 24125 Bergamo, Italy;
| | | | - Sergio Bracarda
- Department of Medical Oncology, "Santa Maria" Hospital, 05100 Terni, Italy;
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-oncology, University of Milan, Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, 20132 Milan, Italy;
| | - Luigi Da Pozzo
- Department of Urology, Università degli Studi Milano Bicocca - ASST Papa Giovanni XXIII, 24129 Bergamo, Italy;
| | - Giario Natale Conti
- Urology Unit, Azienda Socio-Sanitaria Territoriale Lariana, Sant’Anna Hospital, 22042 Como, Italy;
| |
Collapse
|
61
|
Dirix PR, Mercier C, Dirix LY. Re: Fabian Lohaus, Klaus Zöphel, Steffen Löck, et al. Can Local Ablative Radiotherapy Revert Castration-resistant Prostate Cancer to an Earlier Stage of Disease? Eur Urol 2019;75:548–51. Eur Urol 2019; 76:e101-e102. [DOI: 10.1016/j.eururo.2019.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/18/2019] [Indexed: 11/29/2022]
|
62
|
Wülfing C, Bögemann M, Goebell PJ, Hammerer P, Machtens S, Pfister D, Schwentner C, Steuber T, von Amsberg G, Schostak M. [Treatment situation in metastastic Castration Naive Prostate Cancer (mCRPC) and the implications on clinical routine]. Urologe A 2019; 58:1066-1072. [PMID: 31041460 DOI: 10.1007/s00120-019-0925-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is an ongoing change of paradigm in the treatment of metastatic prostate cancer (mPC). Taxan-based chemotherapy demonstrated a prolonged survival of patients in several randomized phase III trials. This is true in the situation of metastatic castration-resistent prostate cancer (mCRPC) as well as in the hormone-naïve stage (metastatic castration-naive PC [mCNPC]). In patients with mCNPC, treatment with docetaxel in combination with androgen deprivation therapy (ADT) prolonged the median total survival time by 15 months in comparison to ADT alone. Comparable results were obtained by the endocrine combination treatment with ADT/abiraterone. With the current data in mind it seems to be useful to discuss the value of early combination therapy with ADT/docetaxel or ADT/abiraterone as well as the impact on further treatment options in the mCRPC setting and to define criteria for treatment decisions in clinical practice.
Collapse
Affiliation(s)
- C Wülfing
- Abteilung für Urologie, Asklepios Klinik Altona, Paul-Ehrlich-Straße 1, 22763, Hamburg, Deutschland.
| | - M Bögemann
- Klinik für Urologie und Kinderurologie, Universitätsklinikum Münster, Münster, Deutschland
| | - P J Goebell
- Urologische und Kinderurologische Klinik, Universitätsklinikum Erlangen, Erlangen, Deutschland
| | - P Hammerer
- Klinik für Urologie und Uro-Onkologie, Städtisches Klinikum Braunschweig, Braunschweig, Deutschland
| | - S Machtens
- Klinik für Urologie und Kinderurologie, GFO Kliniken Rhein Berg, Betriebsstätte, Marien-Krankenhaus Bergisch Gladbach, Bergisch Gladbach, Deutschland
| | - D Pfister
- Klinik für Urologie, Uro-Onkologie, Roboter-assistierte und Spezielle Urologische Chirurgie, Universitätsklinikum Köln, Köln, Deutschland
| | - C Schwentner
- Urologische Klinik, Diakonie-Klinikum Stuttgart, Stuttgart, Deutschland
| | - T Steuber
- Martini-Klinik, Prostatakrebszentrum, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
| | - G von Amsberg
- Klinik für Onkologie, Hämatologie und Knochenmarkstransplantation, Onkologisches Zentrum, Universitätsklinikum Hamburg, Hamburg, Deutschland
| | - M Schostak
- Klinik für Urologie und Kinderurologie, Universitätsklinik Magdeburg, Magdeburg, Deutschland
| |
Collapse
|
63
|
Abstract
Oral enzalutamide (Xtandi®), a second generation androgen receptor inhibitor, is indicated for the treatment of castration-resistant prostate cancer (CRPC) in numerous countries worldwide, with specific indications in this patient population varying between individual countries. Based on extensive experience in the clinical trial and/or real-world settings, oral enzalutamide 160 mg once daily is an effective and generally well tolerated treatment in a broad spectrum of patients with CRPC, including in nonmetastatic and metastatic disease and in chemotherapy-naive and -experienced metastatic CRPC. Enzalutamide is an emerging option for the treatment of men with nonmetastatic CRPC who are at high-risk for developing metastatic disease, and remains an important first-line option in chemotherapy-naive or -experienced patients with metastatic CRPC.
Collapse
Affiliation(s)
- Lesley J Scott
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
64
|
Wang Y, Yu B, Zhong F, Guo Q, Li K, Hou Y, Lin N. MRI-based texture analysis of the primary tumor for pre-treatment prediction of bone metastases in prostate cancer. Magn Reson Imaging 2019; 60:76-84. [DOI: 10.1016/j.mri.2019.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/26/2022]
|
65
|
Perez-Lopez R, Tunariu N, Padhani AR, Oyen WJG, Fanti S, Vargas HA, Omlin A, Morris MJ, de Bono J, Koh DM. Imaging Diagnosis and Follow-up of Advanced Prostate Cancer: Clinical Perspectives and State of the Art. Radiology 2019; 292:273-286. [PMID: 31237493 DOI: 10.1148/radiol.2019181931] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The management of advanced prostate cancer has changed substantially with the availability of multiple effective novel treatments, which has led to improved disease survival. In the era of personalized cancer treatments, more precise imaging may help physicians deliver better care. More accurate local staging and earlier detection of metastatic disease, accurate identification of oligometastatic disease, and optimal assessment of treatment response are areas where modern imaging is rapidly evolving and expanding. Next-generation imaging modalities, including whole-body MRI and molecular imaging with combined PET and CT and combined PET and MRI using novel radiopharmaceuticals, create new opportunities for imaging to support and refine management pathways in patients with advanced prostate cancer. This article demonstrates the potential and challenges of applying next-generation imaging to deliver the clinical promise of treatment breakthroughs.
Collapse
Affiliation(s)
- Raquel Perez-Lopez
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Nina Tunariu
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Anwar R Padhani
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Wim J G Oyen
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Stefano Fanti
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Hebert Alberto Vargas
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Aurelius Omlin
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Michael J Morris
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Johann de Bono
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| | - Dow-Mu Koh
- From the Radiomics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain (R.P.L.); Departments of Radiology (N.T., D.M.K.) and Nuclear Medicine (W.J.G.O.), Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM2 5PT, England; Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, England (A.R.P.); Divisions of Radiotherapy and Imaging (W.J.G.O., D.M.K.) and Clinical Studies & Prostate Cancer Targeted Therapy Group (J.d.B.), Institute of Cancer Research, Sutton, England; Departments of Radiology (S.F.) and Genitourinary Oncology Service and Medicine (M.J.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland (H.A.V., A.O.); Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland (H.A.V., A.O.); and Department of Medicine, Weill Cornell Medicine, New York, NY (M.J.M.)
| |
Collapse
|
66
|
Thana M, Wood LA. Followup imaging studies in metastatic castration-resistant prostate cancer: An individualized approach. Can Urol Assoc J 2019; 13:201-202. [PMID: 31199238 DOI: 10.5489/cuaj.6033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Myuran Thana
- Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| | - Lori A Wood
- Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| |
Collapse
|
67
|
Sternberg CN. Enzalutamide, an oral androgen receptor inhibitor for treatment of castration-resistant prostate cancer. Future Oncol 2019; 15:1437-1457. [PMID: 30848157 DOI: 10.2217/fon-2018-0940] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Androgen receptor (AR) signaling is a key pathway in prostate cancer, and patients are initially treated with androgen deprivation therapy. Patients who have stopped responding to androgen deprivation therapy are considered to have castration-resistant prostate cancer (CRPC), which is still dependent on AR signaling. Enzalutamide, an orally available AR inhibitor, was initially approved by the US FDA for the treatment of patients with metastatic CRPC who have previously received docetaxel. The indication was subsequently extended to include all patients with metastatic CRPC, and most recently to include patients with nonmetastatic CRPC. This review summarizes the body of evidence supporting enzalutamide efficacy and safety in CRPC.
Collapse
Affiliation(s)
- Cora N Sternberg
- Division of Medical Oncology, Weill Cornell Medical Center, New York, NY 10021, USA
| |
Collapse
|
68
|
Petralia G, Padhani AR. Whole-Body Magnetic Resonance Imaging in Oncology: Uses and Indications. Magn Reson Imaging Clin N Am 2019; 26:495-507. [PMID: 30316463 DOI: 10.1016/j.mric.2018.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Whole-body MRI (WB-MRI) has emerged as a radiation-free method for the diagnosis, staging, and therapy response assessments in cancer patients. This article reviews the current roles for WB-MRI in the clinical context of limitations of currently used techniques, focusing on bone marrow disease applications. Indication for broader clinical use are discussed, including guideline recommendations. The emerging screening role of WB-MRI in subjects at high risk of cancer is discussed, as is normal population screening.
Collapse
Affiliation(s)
- Giuseppe Petralia
- Department of Radiology, IEO - European Institute of Oncology IRCCS, Via Ripamonti, 435, Milan 20141, Italy; Department of Oncology and Hematology, University of Milan, Via Festa del Perdono 7, Milan 20122, Italy.
| | - Anwar R Padhani
- MR unit, Paul Strickland Scanner Centre, Mount Vernon Hospital, Rickmansworth Road, Northwood, Middlesex HA6 2RN, UK
| |
Collapse
|
69
|
Prognostic Association of Prostate-specific Antigen Decline with Clinical Outcomes in Men with Metastatic Castration-resistant Prostate Cancer Treated with Enzalutamide in a Randomized Clinical Trial. Eur Urol Oncol 2018; 2:677-684. [PMID: 31274110 DOI: 10.1016/j.euo.2018.11.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/18/2018] [Accepted: 11/13/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND In the PREVAIL study, enzalutamide provided significant improvements versus placebo in clinical outcomes in chemotherapy-naïve men with metastatic castration-resistant prostate cancer (mCRPC). The association of post-treatment prostate-specific antigen (PSA) decline with clinical outcomes may provide important prognostic information. OBJECTIVE To evaluate associations between the magnitude of PSA decline from baseline to month 3 and clinical outcomes among enzalutamide recipients. DESIGN, SETTING, AND PARTICIPANTS This was a post hoc retrospective analysis of PREVAIL, an international, randomized, double-blind, placebo-controlled phase 3 study. Men with mCRPC and no prior chemotherapy from the enzalutamide arm were included (n=872). Patients were grouped by confirmed maximal PSA decline from baseline to month 3 of treatment (n=795 evaluable). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Primary outcomes were overall survival and radiographic progression-free survival. Secondary outcomes included PSA progression-free survival, radiographic response, and degradation of Functional Assessment of Cancer Therapy-Prostate score, which were estimated using the Kaplan-Meier method. RESULTS AND LIMITATIONS Following 3mo of enzalutamide treatment, 88% (701/795), 80% (639/795), and 39% (307/795) of patients had postbaseline confirmed maximal PSA declines of ≥30%, ≥50%, and ≥90%, respectively, whereas 12% (94/795) had no confirmed maximal PSA decline or a decline of <30%. Greater degrees of PSA decline within the first 3mo of enzalutamide treatment were increasingly associated with longer overall survival, time to PSA and radiographic progression, higher objective soft-tissue responses, and longer time to quality-of-life deterioration than no PSA decline or declines of <30% from baseline. PSA flares (rise followed by fall) after enzalutamide treatment were rare (<1%). CONCLUSIONS The magnitude of PSA decline after 3mo of enzalutamide therapy was strongly associated with better clinical and patient-reported outcomes. This updated prognostic information is of clinical value to this patient population and their health care providers. PATIENT SUMMARY We report that decreases in PSA levels are closely linked to better health and survival after 3mo of enzalutamide treatment in men with metastatic prostate cancer. The PREVAIL trial is registered at clinicaltrials.gov as NCT01212991.
Collapse
|
70
|
Gómez-Veiga F, Álvarez-Ossorio JL, Carballido-Rodríguez J, Juárez-Soto A, Rodríguez-Antolín A, Cozar-Olmo JM. Radium-223 for the treatment of metastatic castration-resistant prostate cancer: A window of opportunity. Actas Urol Esp 2018; 42:616-624. [PMID: 30041891 DOI: 10.1016/j.acuro.2018.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 11/30/2022]
Abstract
CONTEXT The elimination of bone metastases, restoration and/or preservation of bone morphology and prevention and/or delay of skeletal events are a fundamental objective in the management of metastatic castration-resistant prostate cancer (mCRPC). Radium-223 is the first targeted alpha therapy with effects on bone that has been shown to increase survival in these patients, besides providing other bone-related benefits. OBJECTIVE To analyze the impact of bone metastasis on mCRPC, and the benefits and the window of opportunity provided by radium-223 in the treatment of patients with mCRPC in the current treatment era. EVIDENCE ACQUISITION A bibliographic search of PubMed and Spanish and international congresses on radium-223 and other first-line treatments for mCRPC was performed. Recent guidelines and recommendations by experts were also consulted. SUMMARY OF THE EVIDENCE Evidence for the mechanism of action of radium-223 widen its effects to the tumor bone environment. Survival of patients treated with radium-223 is higher in those with mild symptoms as opposed to those with moderate-severe symptoms. The presence of visceral metastases even in the early stages of mCRPC supports starting radium-223 therapy before the symptoms become clinically relevant. A 3-year study has confirmed its good safety profile. Changes in tALP and LDH may be useful markers for monitoring the treatment with radium-223, but they are not predictors of overall survival. CONCLUSION Radium-223 is a valuable therapeutic alternative in the treatment of patients with mCRPC in early stages of the disease, with a good safety profile. Its benefits extend to the bone environment.
Collapse
Affiliation(s)
- F Gómez-Veiga
- Hospital Clínico Universitario de Salamanca, Salamanca, España.
| | | | | | - A Juárez-Soto
- Hospital de Jerez, Jerez de la Frontera, Cádiz, España
| | | | | |
Collapse
|
71
|
Martin LJ, Alibhai SMH, Komisarenko M, Timilshina N, Finelli A. Identification of subgroups of metastatic castrate-resistant prostate cancer (mCRPC) patients treated with abiraterone plus prednisone at low- vs. high-risk of radiographic progression: An analysis of COU-AA-302. Can Urol Assoc J 2018; 13:192-200. [PMID: 30407155 DOI: 10.5489/cuaj.5586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Radiographic imaging is used to monitor disease progression for men with metastatic castrate-resistant prostate cancer (mCRPC). The optimal frequency of imaging, a costly and limited resource, is not known. Our objective was to identify predictors of radiographic progression to inform the frequency of imaging for men with mCRPC. METHODS We accessed data for men with chemotherapy-naive mCRPC in the abiraterone acetate plus prednisone (AA-P) group of a randomized trial (COU-AA-302) (n=546). We used Cox proportional hazards modelling to identify predictors of time to progression. We divided patients into groups based on the most important predictors and estimated the probability of radiographic progression-free survival (RPFS) at six and 12 months. RESULTS Baseline disease and change in prostate-specific antigen (PSA) at eight weeks were the strongest determinants of RPFS. The probability of RPFS for men with bone-only disease and a ≥50% fall in PSA was 93% (95% confidence interval [CI] 87-96) at six months and 80% (95% CI 72-86) at 12 months. In contrast, the probability of RPFS for men with bone and soft tissue metastasis and <50% fall in PSA was 55% (95% CI 41-67) at six months and 34% (95% CI 22-47) at 12 months. These findings should be externally validated. CONCLUSIONS Patients with chemotherapy-naive mCRPC treated with first-line AA-P can be divided into groups with significantly different risks of radiographic progression based on a few clinically available variables, suggesting that imaging schedules could be individualized.
Collapse
Affiliation(s)
- Lisa J Martin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Shabbir M H Alibhai
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Maria Komisarenko
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Antonio Finelli
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
72
|
Fettke H, Kwan EM, Azad AA. Cell-free DNA in cancer: current insights. Cell Oncol (Dordr) 2018; 42:13-28. [PMID: 30367445 DOI: 10.1007/s13402-018-0413-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The field of liquid biopsies in oncology is rapidly expanding, with the application of cell-free circulating tumour DNA (ctDNA) showing promise in this era of precision medicine. Compared with traditional clinical and radiographic tumour monitoring methods, the analysis of ctDNA provides a minimally-invasive and technically feasible approach to assess temporal and spatial molecular evolutions of the tumour landscape. The constantly advancing technological platforms available for ctDNA extraction and analysis allow greater analytical sensitivities than ever before. The potential translational impact of ctDNA as a blood-based biomarker for the identification, characterization and monitoring of cancer has been demonstrated in numerous proof-of-concept studies, with ctDNA analysis beginning to be applied clinically across multiple facets of oncology. CONCLUSIONS In this review we discuss the biology, recent advancements, technical considerations and clinical implications of ctDNA in the context of cancer, and highlight important challenges and future directions for the integration of ctDNA into standardised patient care.
Collapse
Affiliation(s)
- Heidi Fettke
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia.
| | - Edmond M Kwan
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia.,Department of Medical Oncology, Monash Health, Melbourne, Australia
| | - Arun A Azad
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia.,Department of Medical Oncology, Monash Health, Melbourne, Australia
| |
Collapse
|
73
|
Matsubara N, Yamada Y, Tabata KI, Satoh T, Kamiya N, Suzuki H, Kawahara T, Uemura H, Yano A, Kawakami S, Otsuka M, Fukasawa S. Abiraterone Followed by Enzalutamide Versus Enzalutamide Followed by Abiraterone in Chemotherapy-naive Patients With Metastatic Castration-resistant Prostate Cancer. Clin Genitourin Cancer 2018; 16:142-148. [DOI: 10.1016/j.clgc.2017.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/14/2017] [Accepted: 09/17/2017] [Indexed: 01/05/2023]
|
74
|
Gillessen S, Attard G, Beer TM, Beltran H, Bossi A, Bristow R, Carver B, Castellano D, Chung BH, Clarke N, Daugaard G, Davis ID, de Bono J, Borges Dos Reis R, Drake CG, Eeles R, Efstathiou E, Evans CP, Fanti S, Feng F, Fizazi K, Frydenberg M, Gleave M, Halabi S, Heidenreich A, Higano CS, James N, Kantoff P, Kellokumpu-Lehtinen PL, Khauli RB, Kramer G, Logothetis C, Maluf F, Morgans AK, Morris MJ, Mottet N, Murthy V, Oh W, Ost P, Padhani AR, Parker C, Pritchard CC, Roach M, Rubin MA, Ryan C, Saad F, Sartor O, Scher H, Sella A, Shore N, Smith M, Soule H, Sternberg CN, Suzuki H, Sweeney C, Sydes MR, Tannock I, Tombal B, Valdagni R, Wiegel T, Omlin A. Management of Patients with Advanced Prostate Cancer: The Report of the Advanced Prostate Cancer Consensus Conference APCCC 2017. Eur Urol 2018; 73:178-211. [PMID: 28655541 DOI: 10.1016/j.eururo.2017.06.002] [Citation(s) in RCA: 369] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND In advanced prostate cancer (APC), successful drug development as well as advances in imaging and molecular characterisation have resulted in multiple areas where there is lack of evidence or low level of evidence. The Advanced Prostate Cancer Consensus Conference (APCCC) 2017 addressed some of these topics. OBJECTIVE To present the report of APCCC 2017. DESIGN, SETTING, AND PARTICIPANTS Ten important areas of controversy in APC management were identified: high-risk localised and locally advanced prostate cancer; "oligometastatic" prostate cancer; castration-naïve and castration-resistant prostate cancer; the role of imaging in APC; osteoclast-targeted therapy; molecular characterisation of blood and tissue; genetic counselling/testing; side effects of systemic treatment(s); global access to prostate cancer drugs. A panel of 60 international prostate cancer experts developed the program and the consensus questions. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The panel voted publicly but anonymously on 150 predefined questions, which have been developed following a modified Delphi process. RESULTS AND LIMITATIONS Voting is based on panellist opinion, and thus is not based on a standard literature review or meta-analysis. The outcomes of the voting had varying degrees of support, as reflected in the wording of this article, as well as in the detailed voting results recorded in Supplementary data. CONCLUSIONS The presented expert voting results can be used for support in areas of management of men with APC where there is no high-level evidence, but individualised treatment decisions should as always be based on all of the data available, including disease extent and location, prior therapies regardless of type, host factors including comorbidities, as well as patient preferences, current and emerging evidence, and logistical and economic constraints. Inclusion of men with APC in clinical trials should be strongly encouraged. Importantly, APCCC 2017 again identified important areas in need of trials specifically designed to address them. PATIENT SUMMARY The second Advanced Prostate Cancer Consensus Conference APCCC 2017 did provide a forum for discussion and debates on current treatment options for men with advanced prostate cancer. The aim of the conference is to bring the expertise of world experts to care givers around the world who see less patients with prostate cancer. The conference concluded with a discussion and voting of the expert panel on predefined consensus questions, targeting areas of primary clinical relevance. The results of these expert opinion votes are embedded in the clinical context of current treatment of men with advanced prostate cancer and provide a practical guide to clinicians to assist in the discussions with men with prostate cancer as part of a shared and multidisciplinary decision-making process.
Collapse
Affiliation(s)
- Silke Gillessen
- Department of Medical Oncology, Cantonal Hospital St. Gallen and University of Berne, Switzerland.
| | - Gerhardt Attard
- Department of Medical Oncology, The Institute of Cancer Research/Royal Marsden, London, UK
| | - Tomasz M Beer
- Oregon Health & Science University Knight Cancer Institute, OR, USA
| | - Himisha Beltran
- Department of Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Alberto Bossi
- Department of Radiation Oncology, Genito Urinary Oncology, Prostate Brachytherapy Unit, Goustave Roussy, Paris, France
| | - Rob Bristow
- Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, USA
| | - Brett Carver
- Department of Urology, Sidney Kimmel Center for Prostate and Urologic Cancers, New York, NY, USA
| | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Byung Ha Chung
- Department of Urology, Gangnam Severance Hospital, Yonsei University Health System, Seoul, Korea
| | - Noel Clarke
- Department of Urology, The Christie and Salford Royal Hospitals, Manchester, UK
| | - Gedske Daugaard
- Department of Medical Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ian D Davis
- Monash University and Eastern Health, Eastern Health Clinical School, Box Hill, Australia
| | - Johann de Bono
- Department of Medical Oncology, The Institute of Cancer Research/Royal Marsden, London, UK
| | - Rodolfo Borges Dos Reis
- Department of Urology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Charles G Drake
- Department of Medical Oncology, Division of Haematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | - Ros Eeles
- Department of Clinical Oncology and Genetics, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | - Eleni Efstathiou
- Department of Medical Oncology, University of Texas MD Anderson Cancer Center, TX, USA
| | - Christopher P Evans
- Department of Urology, University of California, Davis School of Medicine, CA, USA
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, Università di Bologna, Italy
| | - Felix Feng
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Karim Fizazi
- Department of Medical Oncology, Gustave Roussy, University of Paris Sud, Paris, France
| | - Mark Frydenberg
- Department of Surgery, Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University
| | - Martin Gleave
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Susan Halabi
- Department of Clinical trials and Statistics, Duke University, Durham, NC, USA
| | | | - Celestia S Higano
- Department of Medicine, Division of Medical Oncology, University of Washington and Fred Hutchinson Cancer Research Center, WA, USA
| | - Nicolas James
- Department of Clinical Oncology, Clinical Oncology Queen Elizabeth Hospital Birmingham and University of Birmingham, Birmingham, UK
| | - Philip Kantoff
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Pirkko-Liisa Kellokumpu-Lehtinen
- Department of Clinical Oncology, Tampere University Hospital, Faculty of Medicine and Life Sciences, University of Tampere, Finland
| | - Raja B Khauli
- Department of Urology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Chris Logothetis
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Centre, Houston, TX, USA
| | - Fernando Maluf
- Department of Medical Oncology Hospital Israelita Albert Einstein and Department of Medical Oncology Beneficência Portuguesa de São Paulo
| | - Alicia K Morgans
- Department of Medical Oncology and Epidemiology, Vanderbilt University Medical Center, Division of Hematology/Oncology, Nashville, TN, USA
| | - Michael J Morris
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicolas Mottet
- Department of Urology, University Hospital Nord St. Etienne, St. Etienne, France
| | - Vedang Murthy
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, India
| | - William Oh
- Department of Medical Oncology, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| | - Piet Ost
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Anwar R Padhani
- Department of Radiology, Mount Vernon Cancer Centre and Institute of Cancer Research, London, UK
| | - Chris Parker
- Department of Clinical Oncology, Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Mack Roach
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Mark A Rubin
- Department of Pathology, University of Bern and the Inselspital, Bern (CH)
| | - Charles Ryan
- Department of Medical Oncology, Clinical Medicine and Urology at the Helen Diller Family Comprehensive Cancer Center at the University of, California, San Francisco, CA, USA
| | - Fred Saad
- Department of Urology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Oliver Sartor
- Department of Medical Oncology, Tulane Cancer Center, New Orleans, LA, USA
| | - Howard Scher
- Department of Medical Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Avishay Sella
- Department of Medical Oncology, Department of Oncology, Assaf Harofeh Medical Centre, Tel-Aviv University, Sackler School of Medicine, Zerifin, Israel
| | - Neal Shore
- Department of Urology, Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Matthew Smith
- Department of Medical Oncology, Massachusetts General Hospital Cancer Centre, Boston, MA, USA
| | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | - Cora N Sternberg
- Department of Medical Oncology, San Camillo Forlanini Hospital, Rome, Italy
| | - Hiroyoshi Suzuki
- Department of Urology, Toho University Sakura Medical Center, Japan
| | - Christopher Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Ian Tannock
- Department of Medical Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Bertrand Tombal
- Department of Urology, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Riccardo Valdagni
- Department of Oncology and Haemato-oncology, Università degli Studi di Milano. Radiation Oncology 1, Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Thomas Wiegel
- Department of Radiation Oncology, Klinik für Strahlentherapie und Radioonkologie des Universitätsklinikum Ulm, Albert-Einstein-Allee, Ulm, Germany
| | - Aurelius Omlin
- Department of Medical Oncology, Cantonal Hospital St. Gallen and University of Berne, Switzerland
| |
Collapse
|