1
|
Gong J, Janes JL, Trustram Eve C, Stock S, Waller J, De Hoedt AM, Kim J, Ghate SR, Shui IM, Freedland SJ. Epidemiology, treatment patterns, and clinical outcomes in de novo oligometastatic hormone-sensitive prostate cancer. Cancer 2024; 130:3815-3825. [PMID: 38950063 DOI: 10.1002/cncr.35466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND This study was conducted to better characterize the epidemiology, clinical outcomes, and current treatment patterns of de novo oligometastatic hormone-sensitive prostate cancer (omHSPC) in the United States Veterans Affairs Health Care System. METHODS In this observational retrospective cohort study, 400 de novo metastatic hormone-sensitive PC (mHSPC) patients diagnosed from January 2015 to December 2020 (follow-up through December 2021) were randomly selected. omHSPC was defined as five or less total metastases (excluding liver) by conventional imaging. Kaplan-Meier methods estimated overall survival (OS) and castration-resistant prostate cancer (CRPC)-free survival from mHSPC diagnosis date and a log-rank test compared these outcomes by oligometastatic status. RESULTS Twenty percent (79 of 400) of de novo mHSPC patients were oligometastatic. Most baseline characteristics were similar by oligometastatic status; however, men with non-omHSPC had higher median prostate-specific antigen at diagnosis (151.7) than omHSPC (44.1). First-line (1L) novel hormonal therapy was similar between groups (20%); 1L chemotherapy was lower in omHSPC (5%) versus non-omHSPC (14%). More omHSPC patients received metastasis-directed therapy/prostate radiation therapy (14%) versus non-omHSPC (2%). Median OS and CRPC-free survival (in months) were higher in omHSPC versus non-omHSPC (44.4; 95% confidence interval [CI], 33.9-not estimated vs. 26.2; 95% CI, 20.5-32.5, p = .0089 and 27.6; 95% CI, 22.1-37.2 vs. 15.3; 95% CI, 12.8-17.9, p = .0049), respectively. CONCLUSIONS Approximately 20% of de novo mHSPC were oligometastatic, and OS was significantly longer in omHSPC versus non-omHSPC. Although potentially "curative" therapy use was higher in omHSPC versus non-omHSPC, the percentages were still relatively low. Future studies are warranted given potential for prolonged responses with multimodal therapy inclusive of systemic and local therapies.
Collapse
Affiliation(s)
- Jun Gong
- Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jessica L Janes
- Department of Surgery, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | - Claire Trustram Eve
- Department of Surgery, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | - Shannon Stock
- Department of Surgery, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Department of Mathematics and Computer Science, College of the Holy Cross, Worcester, Massachusetts, USA
| | - Justin Waller
- Department of Surgery, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | - Amanda M De Hoedt
- Department of Surgery, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | - Jeri Kim
- Merck & Co., Inc, Rahway, New Jersey, USA
| | | | | | - Stephen J Freedland
- Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| |
Collapse
|
2
|
Stewart TF, Chalfin H, Simon N, Tan A, Apolo A, McKay RR. Perioperative Use of ctDNA to Guide Treatment for Urothelial Carcinoma: The Future is Now. Bladder Cancer 2024; 10:183-198. [PMID: 39493820 PMCID: PMC11530029 DOI: 10.3233/blc-230105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/29/2024] [Indexed: 11/05/2024]
Abstract
Muscle-invasive bladder cancer represents a potentially curable disease, yet often disease recurs and is ultimately fatal. Outcomes for patients with localized urothelial carcinoma are heterogeneous with some patients cured with surgery alone, deriving no benefit from perioperative systemic therapy, while others are left with residual disease and may benefit from additional therapy. Neoadjuvant chemotherapy increases cure rates but comes with significant toxicity. Recently, adjuvant nivolumab has demonstrated significant improvement in disease free survival (DFS), and overall survival analysis is pending. With more therapies approved for urothelial cancer within the last 5 years than ever before, there is incredible potential to improve clinical outcomes and potentially cure more patients with integrated multimodal therapy. Biomarkers are needed to dichotomize those most likely to benefit from perioperative systemic therapy for residual disease, and de-escalate therapy for those likely to be cured with surgery alone. Ultrasensitive assays for circulating tumor DNA (ctDNA) have emerged as a method to identify patients at high risk of recurrence after definitive therapy and may benefit from escalated therapy, while also identifying those least likely to benefit from systemic therapy. Studies have demonstrated that the presence of ctDNA after surgery is prognostic of disease recurrence across multiple cancer types, including bladder cancer, but questions remain as to the utility of these tests, and whether they can be predictive of benefit of adjuvant therapy. Although these liquid biopsies hold significant promise to transform perioperative treatment, prospective studies are needed to validate their utility as prognostic and predictive biomarkers. To bridge this knowledge gap, contemporary clinical trials are incorporating ctDNA as an integral biomarker to guide therapy for MIBC.
Collapse
Affiliation(s)
- Tyler F. Stewart
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | - Alan Tan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrea Apolo
- National Cancer Institute, NIH, Bethesda, MD, USA
| | - Rana R. McKay
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
3
|
Giunta EF, Roviello G, Conteduca V, Verzoni E, Procopio G, De Giorgi U. Pharmacological treatment landscape of non-metastatic hormone-sensitive prostate cancer: a narrative review on behalf of the Meet-URO Group. Crit Rev Oncol Hematol 2024:104534. [PMID: 39447667 DOI: 10.1016/j.critrevonc.2024.104534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/20/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
The definition of "non-metastatic hormone-sensitive prostate cancer" (nmHSPC) can be applied to patients with prostate cancer (PC) who are androgen-deprivation therapy-naïve and without evidence of metastatic disease. This definition includes heterogeneous situations; however, PC patients at high risk of metastatic spread - and who have not started a hormonal treatment - constitute a unique category with unmet clinical needs. This narrative review critically discusses the advances that characterize the rapidly evolving diagnostic and therapeutic scenario in the nmHSPC setting. We found that nmHSPC represents a grey zone in the context of PC. New clinical trials are trying to redefine the therapeutic algorithm of these patients, but escalating treatment seems not to be the right choice for the overall population. Biomarkers able to stratify patients - including molecular ones - are urgently needed, and biomarker-based clinical trials could clarify their prognostic and predictive role in the nmHSPC scenario.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze, Italy
| | - Vincenza Conteduca
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Elena Verzoni
- SSD Genitourinary Medical Oncology and Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Procopio
- SSD Genitourinary Medical Oncology and Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy.
| |
Collapse
|
4
|
Gillessen S, Turco F, Davis ID, Efstathiou JA, Fizazi K, James ND, Shore N, Small E, Smith M, Sweeney CJ, Tombal B, Zilli T, Agarwal N, Antonarakis ES, Aparicio A, Armstrong AJ, Bastos DA, Attard G, Axcrona K, Ayadi M, Beltran H, Bjartell A, Blanchard P, Bourlon MT, Briganti A, Bulbul M, Buttigliero C, Caffo O, Castellano D, Castro E, Cheng HH, Chi KN, Clarke CS, Clarke N, de Bono JS, De Santis M, Duran I, Efstathiou E, Ekeke ON, El Nahas TIH, Emmett L, Fanti S, Fatiregun OA, Feng FY, Fong PCC, Fonteyne V, Fossati N, George DJ, Gleave ME, Gravis G, Halabi S, Heinrich D, Herrmann K, Hofman MS, Hope TA, Horvath LG, Hussain MHA, Jereczek-Fossa BA, Jones RJ, Joshua AM, Kanesvaran R, Keizman D, Khauli RB, Kramer G, Loeb S, Mahal BA, Maluf FC, Mateo J, Matheson D, Matikainen MP, McDermott R, McKay RR, Mehra N, Merseburger AS, Morgans AK, Morris MJ, Mrabti H, Mukherji D, Murphy DG, Murthy V, Mutambirwa SBA, Nguyen PL, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Parker C, Poon DMC, Pritchard CC, Rabah DM, Rathkopf D, Reiter RE, Renard-Penna R, Ryan CJ, Saad F, Sade JP, Sandhu S, Sartor OA, Schaeffer E, Scher HI, Sharifi N, Skoneczna IA, Soule HR, Spratt DE, Srinivas S, Sternberg CN, Suzuki H, Taplin ME, Thellenberg-Karlsson C, Tilki D, Türkeri LN, Uemura H, Ürün Y, Vale CL, Vapiwala N, Walz J, Yamoah K, Ye D, Yu EY, Zapatero A, Omlin A. Management of Patients with Advanced Prostate Cancer. Report from the 2024 Advanced Prostate Cancer Consensus Conference (APCCC). Eur Urol 2024:S0302-2838(24)02610-1. [PMID: 39394013 DOI: 10.1016/j.eururo.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Innovations have improved outcomes in advanced prostate cancer (PC). Nonetheless, we continue to lack high-level evidence on a variety of topics that greatly impact daily practice. The 2024 Advanced Prostate Cancer Consensus Conference (APCCC) surveyed experts on key questions in clinical management in order to supplement evidence-based guidelines. Here we present voting results for questions from APCCC 2024. METHODS Before the conference, a panel of 120 international PC experts used a modified Delphi process to develop 183 multiple-choice consensus questions on eight different topics. Before the conference, these questions were administered via a web-based survey to the voting panel members ("panellists"). KEY FINDINGS AND LIMITATIONS Consensus was a priori defined as ≥75% agreement, with strong consensus defined as ≥90% agreement. The voting results show varying degrees of consensus, as discussed in this article and detailed in the Supplementary material. These findings do not include a formal literature review or meta-analysis. CONCLUSIONS AND CLINICAL IMPLICATIONS The voting results can help physicians and patients navigate controversial areas of clinical management for which high-level evidence is scant or conflicting. The findings can also help funders and policymakers in prioritising areas for future research. Diagnostic and treatment decisions should always be individualised on the basis of patient and cancer characteristics, and should incorporate current and emerging clinical evidence, guidelines, and logistic and economic factors. Enrolment in clinical trials is always strongly encouraged. Importantly, APCCC 2024 once again identified important gaps (areas of nonconsensus) that merit evaluation in specifically designed trials.
Collapse
Affiliation(s)
- Silke Gillessen
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland.
| | - Fabio Turco
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Ian D Davis
- Monash University, Melbourne, Australia; Eastern Health, Melbourne, Australia
| | | | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | | | - Neal Shore
- Carolina Urologic Research Center and GenesisCare, Myrtle Beach, SC, USA
| | - Eric Small
- 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
| | - Christopher J Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia
| | - Bertrand Tombal
- Division of Urology, Clinique Universitaire St. Luc, Brussels, Belgium
| | - Thomas Zilli
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Armstrong
- Center for Prostate and Urologic Cancer, Duke Cancer Institute, Duke University, Durham, NC, USA
| | | | | | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | - Mouna Ayadi
- Salah Azaiz Institute, Medical School of Tunis, Tunis, Tunisia
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Anders Bjartell
- Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Pierre Blanchard
- Department of Radiation Oncology, Oncostat U1018 INSERM, Université Paris-Saclay, Gustave-Roussy, Villejuif, France
| | - Maria T Bourlon
- Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Muhammad Bulbul
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Consuelo Buttigliero
- Department of Oncology, San Luigi Hospital, University of Turin, Orbassano, Italy
| | - Orazio Caffo
- Medical Oncology Department, Santa Chiara Hospital, APSS, Trento, Italy
| | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Heather H Cheng
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Kim N Chi
- BC Cancer and University of British Columbia, Vancouver, Canada
| | - Caroline S Clarke
- Research Department of Primary Care and Population Health, University College London, London, UK
| | - Noel Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - Johann S de Bono
- Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Ignacio Duran
- Medical Oncology Department, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Spain
| | | | - Onyeanunam N Ekeke
- Urology Division, University of Port Harcourt Teaching Hospital, Port Harcourt, Nigeria
| | | | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia; Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Stefano Fanti
- Department of Nuclear Medicine, IRCCS AOU Bologna, Bologna, Italy
| | | | - Felix Y Feng
- University of California-San Francisco, San Francisco, CA, USA
| | - Peter C C Fong
- Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | | | - Nicola Fossati
- Department of Surgery (Urology Service), Ente Ospedaliero Cantonale, Università della Svizzera Italiana Lugano, Switzerland
| | - Daniel J George
- Departments of Medicine and Surgery, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Martin E Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Daniel Heinrich
- Department of Oncology and Radiotherapy, Innlandet Hospital Trust, Gjøvik, Norway
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium, University Hospital Essen, Essen, Germany
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California-San Francisco, San Francisco, CA, USA
| | - Lisa G Horvath
- Chris O'Brien Lifehouse, University of Sydney, Sydney, Australia
| | - Maha H A Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy; Department of Radiation Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Robert J Jones
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Anthony M Joshua
- Department of Medical Oncology, Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, Australia
| | | | - Daniel Keizman
- Genitourinary Unit, Division of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Raja B Khauli
- Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon; Division of Urology, Carle-Illinois College of Medicine, Urbana, IL, USA
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Stacy Loeb
- Department of Urology and Population Health, New York University Langone Health, New York, NY, USA; Department of Surgery/Urology, Manhattan Veterans Affairs, New York, NY, USA
| | - Brandon A Mahal
- Department of Radiation Oncology, University of Miami Sylvester Cancer Center, Miami, FL, USA
| | - Fernando C Maluf
- Beneficiência Portuguesa de São Paulo, São Paulo, Brazil; Departamento de Oncologia, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Joaquin Mateo
- Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - David Matheson
- Faculty of Education Health and Wellbeing, University of Wolverhampton, Walsall, UK
| | - Mika P Matikainen
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Ray McDermott
- Department of Medical Oncology, St. Vincent's University Hospital and Cancer Trials, Dublin, Ireland
| | - Rana R McKay
- University of California-San Diego, Palo Alto, CA, USA
| | - Niven Mehra
- Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Alicia K Morgans
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hind Mrabti
- Institut National d'Oncologie, Mohamed V University, Rabat, Morocco
| | - Deborah Mukherji
- Clemenceau Medical Center, Dubai, United Arab Emirates; Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Vedang Murthy
- Radiation Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Shingai B A Mutambirwa
- Department of Urology, Sefako Makgatho Health Science University, Dr. George Mukhari Academic Hospital, Medunsa, South Africa
| | - Paul L Nguyen
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - William K Oh
- Division of Hematology and Medical Oncology, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, Antwerp, Belgium; Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Joe M O'Sullivan
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| | - Chris Parker
- Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Darren M C Poon
- Hong Kong Sanatorium and Hospital, Chinese University of Hong Kong, Hong Kong, China
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Danny M Rabah
- Cancer Research Chair and Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Urology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dana Rathkopf
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Raphaele Renard-Penna
- Department of Imagery, GRC 5 Predictive Onco-Uro, Pitie-Salpetriere Hospital, AP-HP, Sorbonne University, Paris, France
| | - Charles J Ryan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Fred Saad
- Centre Hospitalier de Université de Montréal, Montreal, Canada
| | | | - Shahneen Sandhu
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Oliver A Sartor
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| | - Edward Schaeffer
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nima Sharifi
- Desai Sethi Urology Institute and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Iwona A Skoneczna
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Sandy Srinivas
- Division of Medical Oncology, Stanford University Medical Center, Stanford, CA, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Division of Hematology and Oncology, Meyer Cancer Center, New York Presbyterian Hospital, New York, NY, USA
| | - Hiroyoshi Suzuki
- Department of Urology, Toho University Sakura Medical Center, Sakura, Japan
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Derya Tilki
- Martini-Klinik Prostate Cancer Center and Department of Urology, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Levent N Türkeri
- Department of Urology, M.A. Aydınlar Acıbadem University, Altunizade Hospital, Istanbul, Turkey
| | - Hiroji Uemura
- Yokohama City University Medical Center, Yokohama, Japan
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University School of Medicine, Ankara, Turkey
| | - Claire L Vale
- MRC Clinical Trials Unit, University College London, London, UK
| | - Neha Vapiwala
- Department of Radiation Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jochen Walz
- Institut Paoli-Calmettes Cancer Center, Marseille, France
| | - Kosj Yamoah
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Evan Y Yu
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Almudena Zapatero
- University Hospital La Princesa, Health Research Institute, Madrid, Spain
| | - Aurelius Omlin
- Onkozentrum Zurich, University of Zurich and Tumorzentrum Hirslanden Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Westhofen T, Feyerabend E, Buchner A, Schlenker B, Becker A, Eismann L, Rodler S, Jokisch F, Stief CG, Kretschmer A. Impact of Preoperative LUTS on Health-related Quality of Life Following Radical Prostatectomy: A Propensity Score Matched Longitudinal Study. Urology 2024; 192:52-58. [PMID: 38679296 DOI: 10.1016/j.urology.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVE To assess the impact of preoperative lower urinary tract symptoms (LUTS) on long-term health-related quality of life (HRQOL) up to 10 years after radical prostatectomy (RP) for prostate cancer (PC). METHODS Within our prospective institutional database of 6487 patients treated with RP for PC (2008-2020), 2727 patients with preoperative LUTS (IPSS score of ≥8) were identified. A 1:1 propensity-score matched analysis of 3056 men (n = 1528 LUTS, n = 1528 no LUTS) was conducted. Primary endpoint was HRQOL (based on EORTC QLQ-C30 and PR25). Linear regression models tested the effect of preoperative LUTS on the net change in general HRQOL (P <.05). RESULTS Median follow-up was 48 months. Preoperative mean global health status (GHS) score (67.4 vs 75.7) was significantly lower in the LUTS cohort (P <.001). Post-RP the difference in general HRQOL between the LUTS cohort and the no-LUTS cohort became smaller (65.7 vs 67.8), however, remaining statistically significant (P = .037). In long-term follow-up, general HRQOL was comparable between both subcohorts (P-range 0.716-0.876). Multivariable linear regression analysis revealed increased preoperative IPSS as an independent predictor for increased perioperative improvement of IPSS (P <.001) CONCLUSION: For patients undergoing RP, preoperative LUTS were associated with a postoperative improvement of HRQOL outcomes. In long-term follow-up, HRQOL was comparable to patients without preoperative LUTS. Hence, RP is an efficient option to treat PC as well as LUTS in those patients.
Collapse
Affiliation(s)
- Thilo Westhofen
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Enya Feyerabend
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Buchner
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Boris Schlenker
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Armin Becker
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lennert Eismann
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Severin Rodler
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Friedrich Jokisch
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian G Stief
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexander Kretschmer
- Department of Urology, University Hospital Großhadern, Ludwig-Maximilians-University Munich, Munich, Germany; Janssen Global Research and Development, Los Angeles, CA
| |
Collapse
|
6
|
Cao Y, Sutera P, Silva Mendes W, Yousefi B, Hrinivich T, Deek M, Phillips R, Song D, Kiess A, Cem Guler O, Torun N, Reyhan M, Sawant A, Marchionni L, Simone NL, Tran P, Onal C, Ren L. Machine learning predicts conventional imaging metastasis-free survival (MFS) for oligometastatic castration-sensitive prostate cancer (omCSPC) using prostate-specific membrane antigen (PSMA) PET radiomics. Radiother Oncol 2024; 199:110443. [PMID: 39094629 PMCID: PMC11405100 DOI: 10.1016/j.radonc.2024.110443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE This study investigated imaging biomarkers derived from PSMA-PET acquired pre- and post-metastasis-directed therapy (MDT) to predict 2-year metastasis-free survival (MFS), which provides valuable early response assessment to improve patient outcomes. MATERIALS/METHODS An international cohort of 117 oligometastatic castration-sensitive prostate cancer (omCSPC) patients, comprising 34 from John Hopkins Hospital (JHH) and 83 from Baskent University (BU), were treated with stereotactic ablative radiation therapy (SABR) MDT with both pre- and post-MDT PSMA-PET/CT scans acquired. PET radiomic features were analyzed from a CT-PET fusion defined gross tumor volume ((GTV) or zone 1), and a 5 mm expansion ring area outside the GTV (zone 2). A total of 1748 PET radiomic features were extracted from these zones. The six most significant features selected using the Chi2 method, along with five clinical parameters (age, Gleason score, number of total lesions, untreated lesions, and pre-MDT prostate-specific antigen (PSA)) were extracted as inputs to the models. Various machine learning models, including Random Forest, Decision Tree, Support Vector Machine, and Naïve Bayesian, were employed for 2-year MFS prediction and tested using leave-one-out and cross-institution validation. RESULTS Six radiomic features, including Total Energy, Entropy, and Standard Deviation from pre-PSMA-PET zone 1, Total Energy and Contrast from post-PSMA-PET zone 1, and Entropy from pre-PSMA-PET zone 2, along with five clinical parameters were selected for predicting 2-year MFS. In a leave-one-out test with all the patients, random forest achieved an accuracy of 80 % and an AUC of 0.82 in predicting 2-year MFS. In cross-institution validation, the model correctly predicted 2-year MFS events with an accuracy of 75 % and an AUC of 0.77 for patients from JHH, and an accuracy of 78 % and an AUC of 0.80 for BU patients, respectively. CONCLUSION Our study demonstrated the promise of using pre- and post-MDT PSMA-PET-based imaging biomarkers for MFS prediction for omCSPC patients.
Collapse
Affiliation(s)
- Yufeng Cao
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William Silva Mendes
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bardia Yousefi
- Fischell Department of Bioengineering, University of Maryland School of Medicine, College Park, MD, USA
| | - Tom Hrinivich
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Ryan Phillips
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Danny Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ana Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ozan Cem Guler
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Adana, Turkey
| | - Nese Torun
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Nuclear Medicine, Ankara, Turkey
| | - Mehmet Reyhan
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Nuclear Medicine, Ankara, Turkey
| | - Amit Sawant
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nicole L Simone
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Phuoc Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Cem Onal
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Adana, Turkey; Baskent University Faculty of Medicine, Department of Radiation Oncology, Ankara, Turkey.
| | - Lei Ren
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Ong WL, Romero T, Roy S, Nikitas J, Joseph D, Zapatero A, Malone S, Morgan SC, Steinberg ML, Valle LF, Zaorsky NG, Martin Ma T, Rettig MB, Nickols N, Jiang T, Reiter RE, Eleswarapu SV, Maldonado X, Sun Y, Nguyen PL, Millar JL, Martin JM, Spratt DE, Kishan AU. Testosterone Recovery Following Androgen Suppression and Prostate Radiotherapy (TRANSPORT): A Pooled Analysis of Five Randomized Trials from the Meta-Analysis of Randomized Trials in Cancer of the Prostate (MARCAP) Consortium. Eur Urol 2024:S0302-2838(24)02601-0. [PMID: 39304428 DOI: 10.1016/j.eururo.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND OBJECTIVE Time to testosterone recovery (TR) following androgen deprivation therapy (ADT) with gonadotropin-releasing hormone agonists varies widely. We evaluate TR kinetics and the oncological impact of an effective castration period in patients receiving definitive radiotherapy and ADT for prostate cancer. METHODS We obtained individual patient data from randomized controlled trials of radiotherapy with ADT and prospectively collected serial testosterone data from the MARCAP Consortium. We estimated the times to noncastrate TR (>1.7 nmol/l) and nonhypogonadal TR (>8.0 nmol/l) were estimated for each prescribed ADT duration, and developed corresponding nomograms. The association between effective castration period and metastasis-free survival (MFS) for any given ADT duration was evaluated via multivariable Cox regression. We conducted cubic spline analyses to assess nonlinear associations. KEY FINDINGS AND LIMITATIONS We included 1444 men from five trials in the analysis, of whom 115 received 4 mo, 880 received 6 mo, 353 received 18 mo, 36 received 28 mo, and 60 received 36 mo of ADT. Times to noncastrate TR and to nonhypogonadal TR varied considerably by ADT duration. Higher baseline testosterone and lower age were associated with a higher likelihood of TR (p < 0.001 for both). Effective castration period was not linearly associated with MFS for any ADT duration on Cox regression. Cubic spline analysis revealed that the optimal effective castration period for an MFS benefit was 10.6 mo for men who received 6 mo of ADT and 18 mo for men who received 18 mo of ADT. CONCLUSIONS AND CLINICAL IMPLICATIONS Time to TR varies according to the ADT duration, baseline testosterone, and age. The relationship between effective castration period and MFS may be nonlinear, with a longer effective castration period being helpful for men receiving 6 mo of ADT.
Collapse
Affiliation(s)
- Wee Loon Ong
- Alfred Health Radiation Oncology, Monash University, Melbourne, Australia
| | - Tahmineh Romero
- Division of General Internal Medicine and Health Services Research, University of California-Los Angeles, Los Angeles, CA, USA
| | - Soumyajit Roy
- Department of Radiation Oncology, Rush University Medical Centre, Chicago, IL, USA
| | - John Nikitas
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - David Joseph
- Department of Medicine and Surgery, University of Western Australia, Perth, Australia
| | - Almudena Zapatero
- Department of Radiation Oncology, Health Research Institute, Hospital Universitario de la Princesa, Madrid, Spain
| | - Shawn Malone
- Department of Radiology, Radiation Oncology and Medical Physics, Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, Canada
| | - Scott C Morgan
- Department of Radiation Oncology, University Hospitals Seidman Cancer Centre, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Michael L Steinberg
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Luca F Valle
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, University Hospitals Seidman Cancer Centre, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ting Martin Ma
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Matthew B Rettig
- Department of Medical Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Nicholas Nickols
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Tommy Jiang
- Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Robert E Reiter
- Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Sriram V Eleswarapu
- Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA
| | | | - Yilun Sun
- Department of Radiation Oncology, University Hospitals Seidman Cancer Centre, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Population Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Paul L Nguyen
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeremy L Millar
- Alfred Health Radiation Oncology, Monash University, Melbourne, Australia
| | - Jarad M Martin
- Department of Radiation Oncology, Calvary Mater Newcastle Hospital, University of Newcastle, Newcastle, Australia
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Centre, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Niazi T, Nabid A, Malagon T, Tisseverasinghe S, Bettahar R, Dahmane R, Martin AG, Jolicoeur M, Yassa M, Barkati M, Igidbashian L, Bahoric B, Archambault R, Villeneuve H, Mohiuddin M. Hypofractionated Dose Escalation Radiotherapy for High-risk Prostate Cancer: The Survival Analysis of the Prostate Cancer Study 5, a Groupe de Radio-oncologie Génito-urinaire du Quebec-led Phase 3 Trial. Eur Urol 2024:S0302-2838(24)02574-0. [PMID: 39271420 DOI: 10.1016/j.eururo.2024.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/24/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND AND OBJECTIVE Prostate Cancer Study 5 (PCS5) compared conventional fractionated radiotherapy (CFRT) with hypofractionated radiotherapy (HFRT) in high-risk prostate cancer (PCa) patients, hypothesizing similar toxicity and survival outcomes. This report presents the efficacy analysis. METHODS PCS5 is a Canadian multicenter, open-label, phase 3 randomized control trial. Men with histologically proven, clinically localized PCa with one or more high-risk features (T3/T4, Gleason score ≥8, and prostate-specific antigen >20) were eligible. Patients were randomized 1:1 to CFRT (76 Gy/38 fractions [Fx] to the prostate and 46 Gy/23 Fx to the pelvic lymph nodes [PLNs]) or HFRT (68 Gy/25 Fx to the prostate and 45 Gy/25 Fx to the PLNs) and 28 mo of androgen suppression. The primary endpoint was toxicity; secondary endpoints included survival outcomes. KEY FINDINGS AND LIMITATIONS Of 329 patients, 164 were randomized to HFRT and 165 to CFRT, with 159 in the HFRT arm and 160 in the CFRT arm included in survival analyses. At the 5-yr median follow-up, there were no significant differences in overall survival (OS; 90.3% vs 89.7%; risk ratio [RR]: 1.01; 95% confidence interval [CI]: 0.93-1.09), PCa-specific survival (PCSS; 97.4% vs 97.5%; RR: 1.00; 95% CI: 0.93-1.07), biochemical recurrence-free survival (BCRFS; 85.2% vs 85.2%; RR: 1.00; 95% CI: 0.91-1.10), or distant metastasis-free survival (DMFS; 87.1% vs 87.1%; RR: 1.00; 95% CI: 0.92-1.09). Hazard ratios were 0.92 (95% CI: 0.56-1.53) for OS, 1.31 (95% CI: 0.46-3.78) for PCSS, 0.85 (95% CI: 0.56-1.30) for BCRFS, and 0.90 (95% CI: 0.56-1.43) for DMFS. Sample size was a limiting factor. CONCLUSIONS AND CLINICAL IMPLICATIONS There were no differences in survival outcomes between HFRT (68 Gy/25 Fx) and CFRT (76 Gy/38 Fx). HFRT, including PLN radiotherapy and long-term androgen deprivation therapy, should be considered a new standard of care for high-risk PCa patients undergoing external beam radiotherapy.
Collapse
Affiliation(s)
- Tamim Niazi
- Jewish General Hospital, McGill University, Montreal, Quebec, Canada.
| | - Abdenour Nabid
- Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
| | - Talia Malagon
- Department of Oncology, McGill University, Montréal, Quebec, Canada; St Mary's Research Centre, Montréal West Island CIUSSS, Montréal, Quebec, Canada
| | | | - Redouane Bettahar
- Centre Hospitalier Régional de Rimouski-Centre de Cancer, Rimouski, Quebec, Canada
| | - Rafika Dahmane
- Pavillon Ste-Marie Centre Hospitalier Régional de Trois-Rivières (CHRTR), Trois-Rivières, Quebec, Canada
| | - Andre-Guy Martin
- Centre Hospitalier Universitaire de Québec (CHUQ)-L'Hôtel-Dieu de Québec, Quebec City, Quebec, Canada
| | | | - Michael Yassa
- Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Maroie Barkati
- Centre Hospitalier de l'Université de Montréal (CHUM) (MB), Montreal, Quebec, Canada
| | | | - Boris Bahoric
- Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | - Md Mohiuddin
- Saint John Regional Hospital (MM), Saint John, New Brunswick, Canada
| |
Collapse
|
9
|
Saripalli AL, Venkatesulu BP, Nickols NG, Valle LF, Harkenrider MM, Kishan AU, Solanki AA. Systematic review and recommendations for re-irradiation for intraprostatic radiorecurrent prostate cancer after definitive radiation therapy. World J Urol 2024; 42:520. [PMID: 39264453 DOI: 10.1007/s00345-024-05205-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 07/17/2024] [Indexed: 09/13/2024] Open
Abstract
PURPOSE Intraprostatic recurrence (IRR) of prostate cancer after radiation therapy is increasingly identified. Our objective was to review the literature to determine the optimal workup for identifying IRR, the management options, and practical considerations for the delivery of re-irradiation as salvage local therapy. METHODS We performed a systematic review of available publications and ongoing studies on the topics of IRR, with a focus on salvage re-irradiation. RESULTS Work up of biochemically recurrent prostate cancer includes PSMA PET/CT and multiparametric MRI, followed by biopsy to confirm IRR. Management options include continued surveillance, palliative hormonal therapy, and salvage local therapy. Salvage local therapy can be delivered using re-irradiation with low dose rate brachytherapy, high dose rate (HDR) brachytherapy, and stereotactic body radiotherapy (SBRT), as well as non-radiation modalities, such as cryotherapy, high-intensity focused ultrasound, irreversible electroporation and radical prostatectomy. Data demonstrate that HDR brachytherapy and SBRT have similar efficacy compared to the other salvage local therapy modalities, while having more favorable side effect profiles. Recommendations for radiation therapy planning and delivery using HDR and SBRT based on the available literature are discussed. CONCLUSION Salvage re-irradiation is safe and effective and should be considered in patients with IRR.
Collapse
Affiliation(s)
- Anjali L Saripalli
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| | - Bhanu Prasad Venkatesulu
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| | - Nicholas G Nickols
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiation Oncology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Luca F Valle
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiation Oncology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Matthew M Harkenrider
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Abhishek A Solanki
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
10
|
Yasar B, Suh YE, Chapman E, Nicholls L, Henderson D, Jones C, Morrison K, Wells E, Henderson J, Meehan C, Sohaib A, Taylor H, Tree A, van As N. Simultaneous Focal Boost With Stereotactic Radiation Therapy for Localized Intermediate- to High-Risk Prostate Cancer: Primary Outcomes of the SPARC Phase 2 Trial. Int J Radiat Oncol Biol Phys 2024; 120:49-58. [PMID: 38499253 DOI: 10.1016/j.ijrobp.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/20/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE Dose-escalated radiation therapy is associated with better biochemical control at the expense of toxicity. Stereotactic body radiation therapy (SBRT) with dose escalation to the dominant intraprostatic lesion (DIL) provides a logical approach to improve outcomes in high-risk disease while limiting toxicity. This study evaluated the toxicity and quality of life (QoL) with CyberKnife-based SBRT and simultaneous integrated boost in localized prostate cancer. METHODS AND MATERIALS Eligible participants included newly diagnosed, biopsy-proven unfavorable intermediate- to high-risk localized prostate cancer (at least 1 of the following: Gleason ≥4+3, magnetic resonance imaging(MRI)-defined T3a N0, prostate-specific antigen ≥20) with up to 2 MRI-identified DILs. Participants received 36.25 Gy in 5 fractions on alternative days with a simultaneous boost to DIL up to 47.5 Gy as allowed by organ-at-risk constraints delivered by CyberKnife. All participants received androgen deprivation therapy. The primary outcome measure was acute grade 2+ genitourinary toxicity. Acute and late genitourinary and gastrointestinal toxicity using Radiation Therapy Oncology Group scoring, biochemical parameters, International Prostate Symptom Score, International Index of Erectile Function 5, and EQ-5D QoL outcomes were assessed. RESULTS Between 2013 and 2023, 20 participants were enrolled with a median follow-up of 30 months. The median D95 dose to DIL was 47.43 Gy. Cumulative acute grade 2+ genitourinary and gastrointestinal toxicity were 25% and 30%, respectively. One patient developed acute grade 3 genitourinary toxicity (5%). There is no late grade 3 genitourinary or gastrointestinal toxicity to date. International Prostate Symptom Score and urinary QoL scores recovered to baseline by 6 months. Patient-reported outcomes showed no significant change in EQ-5D QoL scores at 12 weeks and 1 year. There are no cases of biochemical relapse reported to date. CONCLUSIONS CyberKnife SBRT-delivered dose of 36.25 Gy to the prostate with a simultaneous integrated boost up to 47.5 Gy is well tolerated. Acute and late genitourinary and gastrointestinal toxicity rates are comparable to other contemporary SBRT trials and series with focal boost.
Collapse
Affiliation(s)
- Binnaz Yasar
- Royal Marsden NHS Foundation Trust, London, United Kingdom; Institute of Cancer Research, London, United Kingdom.
| | - Yae-Eun Suh
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Ewan Chapman
- St Bartholomew's Hospital, London, United Kingdom
| | | | - Daniel Henderson
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Caroline Jones
- Leeds Cancer Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Kirsty Morrison
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Emma Wells
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | - Carole Meehan
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Aslam Sohaib
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Helen Taylor
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Alison Tree
- Royal Marsden NHS Foundation Trust, London, United Kingdom; Institute of Cancer Research, London, United Kingdom
| | - Nicholas van As
- Royal Marsden NHS Foundation Trust, London, United Kingdom; Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
11
|
Sayan M, Tilki D, D'Amico AV. Postoperative Management of Prostate Cancer-Optimizing Prostate Cancer Care. JAMA Oncol 2024; 10:1169-1170. [PMID: 38990544 DOI: 10.1001/jamaoncol.2024.1887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
This Viewpoint discusses the end point analyses and results of the RACICALS-RT study.
Collapse
Affiliation(s)
- Mutlay Sayan
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, Massachusetts
| | - Derya Tilki
- Department of Urology and Martini-Klinik Prostate Cancer Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
- Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Anthony V D'Amico
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
12
|
Kahlmeter Brandell J, Valachis A, Ugge H, Smith D, Johansson B. Moderately hypofractionated prostate-only versus whole-pelvis radiotherapy for high-risk prostate cancer: A retrospective real-world single-center cohort study. Clin Transl Radiat Oncol 2024; 48:100846. [PMID: 39258243 PMCID: PMC11384977 DOI: 10.1016/j.ctro.2024.100846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/08/2024] [Accepted: 08/18/2024] [Indexed: 09/12/2024] Open
Abstract
Background The benefit of prophylactic whole pelvis radiation therapy (WPRT) in prostate cancer has been debated for decades, with evidence based mainly on conventional fractionation targeting pelvic nodes. Aim This retrospective cohort study aimed to explore the impact of adding moderately hypofractionated pelvic radiotherapy to prostate-only irradiation (PORT) on prognosis, toxicity, and quality of life in real-world settings. Materials and methods Patients with high-risk and conventionally staged prostate cancer (cT1-3N0M0) treated with moderately hypofractionated WPRT or PORT, using external beam radiotherapy alone or combined with high-dose-rate brachytherapy, at Örebro University Hospital between 2008 and 2021 were identified. Biochemical failure-free survival (BFFS), metastasis-free survival (MFS), prostate cancer-specific survival (PCSS), and overall survival (OS) were compared using Kaplan-Meier method and Cox proportional hazards. Toxicity and quality of life measures were also analysed. Results Among 516 patients (227 PORT, 289 WPRT), 5-year BFFS rates were 77 % (PORT) and 74 % (WPRT), adjusted HR=1.50 (95 % CI=0.88-2.55). No significant differences were found in MFS, PCSS, or OS in main analyses. WPRT was associated with a higher risk of acute grade ≥ 2 and 3 genitourinary toxicities whereas no differences in late toxicities or quality of life between PORT and WPRT were observed. Conclusion We found no significant differences in oncological outcomes or quality of life when comparing moderately hypofractionated PORT to WPRT. Some differences in toxicity patterns were observed. Despite caveats related to study design, our findings support the need for further research on WPRT's impact on treatment-related and patient-reported outcomes.
Collapse
Affiliation(s)
- Jenny Kahlmeter Brandell
- Department of Oncology, Faculty of Medicine and Health, Örebro University Hospital, Örebro University, Örebro, Sweden
| | - Antonis Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University Hospital, Örebro University, Örebro, Sweden
| | - Henrik Ugge
- Department of Urology, Faculty of Medicine and Health, Örebro University Hospital, Örebro University, Örebro, Sweden
| | - Daniel Smith
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden
| | - Bengt Johansson
- Department of Oncology, Faculty of Medicine and Health, Örebro University Hospital, Örebro University, Örebro, Sweden
| |
Collapse
|
13
|
Halabi S, Guo S, Roy A, Rydzewska LE, Godolphin P, Hussain M, Tangen C, Thompson I, Xie W, Carducci MA, Morris MJ, Smith MR, Gravis G, Dearnaley DP, Verhagen PJ, Goto TJ, James ND, Buyse ME, Tierney JF, Sweeney CJ. Reply to J.A. Garcia et al. J Clin Oncol 2024; 42:2940-2941. [PMID: 38875510 PMCID: PMC11346338 DOI: 10.1200/jco.24.00625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 06/16/2024] Open
Affiliation(s)
- Susan Halabi
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Siyuan Guo
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Akash Roy
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Larysa E Rydzewska
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Peter Godolphin
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Maha Hussain
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Catherine Tangen
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Ian Thompson
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Wanling Xie
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Michael A Carducci
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Michael J Morris
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Matthew R Smith
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Gwenaelle Gravis
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - David P Dearnaley
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Paul J Verhagen
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Takayuki J Goto
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Nick D James
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Marc E Buyse
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Jayne F Tierney
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| | - Christopher J Sweeney
- Susan Halabi, PhD, Duke University School of Medicine, Durham, NC; Siyuan Guo, PhD, Duke University School of Medicine, Durham, NC; Akash Roy, PhD, Medical University of South Carolina, Charleston, SC; Larysa E. Rydzewska, PhD and Peter Godolphin, PhD, University College London, London, UK; Maha Hussain, MD, Northwestern University, Chicago, IL; Catherine Tangen, PhD, Fred Hutchinson Cancer Center, Seattle, WA; Ian Thompson, MD, Christus Health, San Antonio, TX; Wanling Xie, MS, Dana Farber Cancer Institute, Boston, MA; Michael A. Carducci, MD, Johns Hopkins University, Baltimore, MD; Michael J. Morris, MD, MSKCC, New York, NY; Matthew R. Smith, MD, PhD, MGH, Boston, MA; Gwenaelle Gravis, MD, Institut Paoli-Calmettes Aix-Mareseille Université, Mareseille, France; David P. Dearnaley, MD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Paul J. Verhagen, MD, Erasmus Medical Center, Rotterdam, the Netherlands; Takayuki J. Goto, MD, Kyoto University, Japan; Nick D. James, MD, PhD, Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK; Marc E. Buyse, PhD, IDDI, Brussels, Belgium; Jayne F. Tierney, PhD, University College London, London, UK; and Christopher J. Sweeney, MD, University of Adelaide, Adelaide, Australia
| |
Collapse
|
14
|
Einstein DJ, Regan MM, Stevens JS, McDermott DF, Madan RA. Metastasis-Free Survival Versus Treatment-Free Survival in Biochemically Recurrent Prostate Cancer: The EMBARK Trial. J Clin Oncol 2024; 42:2849-2852. [PMID: 38754067 PMCID: PMC11457111 DOI: 10.1200/jco.24.00279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/10/2024] [Accepted: 03/25/2024] [Indexed: 05/18/2024] Open
Abstract
What is most important to patients with BCR prostate cancer? Metastasis-free versus treatment-free survival.
Collapse
Affiliation(s)
- David J. Einstein
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Meredith M. Regan
- Harvard Medical School, Boston, MA
- Division of Biostatistics, Dana-Farber Cancer Institute, Boston, MA
| | - Julia S. Stevens
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
| | - David F. McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Ravi A. Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
15
|
Brookman-May SD, Buyse M, Freedland SJ, Miladinovic B, Zhang K, Fendler WP, Feng F, Sartor O, Sweeney CJ. Challenges and Opportunities in Establishing Appropriate Intermediate Endpoints Reflecting Patient Benefit: A Roadmap for Research and Clinical Application in Nonmetastatic Prostate Cancer. Eur Urol 2024; 86:81-87. [PMID: 38762392 DOI: 10.1016/j.eururo.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/01/2024] [Accepted: 04/22/2024] [Indexed: 05/20/2024]
Abstract
Defining meaningful endpoints for research of early-stage high-risk prostate cancer is challenging, with established measures such as overall survival and metastasis-free survival facing limitations related to feasibility and adequate reflection of patient relevance. Developing endpoints must cater to diverse perspectives across scientific, clinical, regulatory, and patient viewpoints. Endpoints such as pathological complete response, no evidence of disease, and prevention of prostate-specific antigen relapse may reflect patient benefit by accounting for diagnostic and treatment burdens.
Collapse
Affiliation(s)
- Sabine D Brookman-May
- Department of Urology, Ludwig-Maximilians University Munich, Munich, Germany; Janssen Research and Development, Spring House, PA, USA.
| | - Marc Buyse
- Data Science Institute, Interuniversity Institute for Biostatistics and statistical Bioinformatics (I-Biostat), University of Hasselt, Hasselt, Belgium; International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Stephen J Freedland
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Section of Urology, Durham VA Medical Center, Durham, NC, USA
| | | | - Ke Zhang
- Janssen Research and Development, San Diego, CA, USA
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Felix Feng
- Department of Medicine, UCSF, San Francisco, CA, USA; Department of Urology, UCSF, San Francisco, CA, USA; Department of Radiation Oncology, UCSF, San Francisco, CA, USA
| | | | - Christopher J Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
16
|
Slevin F, Zattoni F, Checcucci E, Cumberbatch MGK, Nacchia A, Cornford P, Briers E, De Meerleer G, De Santis M, Eberli D, Gandaglia G, Gillessen S, Grivas N, Liew M, Linares Espinós EE, Oldenburg J, Oprea-Lager DE, Ploussard G, Rouvière O, Schoots IG, Smith EJ, Stranne J, Tilki D, Smith CT, Van Den Bergh RCN, Van Oort IM, Wiegel T, Yuan CY, Van den Broeck T, Henry AM. A Systematic Review of the Efficacy and Toxicity of Brachytherapy Boost Combined with External Beam Radiotherapy for Nonmetastatic Prostate Cancer. Eur Urol Oncol 2024; 7:677-696. [PMID: 38151440 DOI: 10.1016/j.euo.2023.11.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023]
Abstract
CONTEXT The optimum use of brachytherapy (BT) combined with external beam radiotherapy (EBRT) for localised/locally advanced prostate cancer (PCa) remains uncertain. OBJECTIVE To perform a systematic review to determine the benefits and harms of EBRT-BT. EVIDENCE ACQUISITION Ovid MEDLINE, Embase, and EBM Reviews-Cochrane Central Register of Controlled Trials databases were systematically searched for studies published between January 1, 2000 and June 7, 2022, according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. Eligible studies compared low- or high-dose-rate EBRT-BT against EBRT ± androgen deprivation therapy (ADT) and/or radical prostatectomy (RP) ± postoperative radiotherapy (RP ± EBRT). The main outcomes were biochemical progression-free survival (bPFS), severe late genitourinary (GU)/gastrointestinal toxicity, metastasis-free survival (MFS), cancer-specific survival (CSS), and overall survival (OS), at/beyond 5 yr. Risk of bias was assessed and confounding assessment was performed. A meta-analysis was performed for randomised controlled trials (RCTs). EVIDENCE SYNTHESIS Seventy-three studies were included (two RCTs, seven prospective studies, and 64 retrospective studies). Most studies included participants with intermediate-or high-risk PCa. Most studies, including both RCTs, used ADT with EBRT-BT. Generally, EBRT-BT was associated with improved bPFS compared with EBRT, but similar MFS, CSS, and OS. A meta-analysis of the two RCTs showed superior bPFS with EBRT-BT (estimated fixed-effect hazard ratio [HR] 0.54 [95% confidence interval {CI} 0.40-0.72], p < 0.001), with absolute improvements in bPFS at 5-6 yr of 4.9-16%. However, no difference was seen for MFS (HR 0.84 [95% CI 0.53-1.28], p = 0.4) or OS (HR 0.87 [95% CI 0.63-1.19], p = 0.4). Fewer studies examined RP ± EBRT. There is an increased risk of severe late GU toxicity, especially with low-dose-rate EBRT-BT, with some evidence of increased prevalence of severe GU toxicity at 5-6 yr of 6.4-7% across the two RCTs. CONCLUSIONS EBRT-BT can be considered for unfavourable intermediate/high-risk localised/locally advanced PCa in patients with good urinary function, although the strength of this recommendation based on the European Association of Urology guideline methodology is weak given that it is based on improvements in biochemical control. PATIENT SUMMARY We found good evidence that radiotherapy combined with brachytherapy keeps prostate cancer controlled for longer, but it could lead to worse urinary side effects than radiotherapy without brachytherapy, and its impact on cancer spread and patient survival is less clear.
Collapse
Affiliation(s)
- Finbar Slevin
- University of Leeds, Leeds, UK; Leeds Cancer Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | - Fabio Zattoni
- Department Surgery, Oncology and Gastroenterology, Urologic Unit, University of Padova, Padova, Italy
| | - Enrico Checcucci
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Hospital, University of Turin, Turin, Italy
| | | | | | - Philip Cornford
- Department of Urology, Liverpool University Hospitals NHS Trust, Liverpool, UK
| | | | - Gert De Meerleer
- Department of Radiotherapy, University Hospitals Leuven, Leuven, Belgium
| | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Daniel Eberli
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Nikolaos Grivas
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Matthew Liew
- Department of Urology, Wrightington, Wigan and Leigh NHS Foundation Trust, Wigan, UK
| | | | - Jan Oldenburg
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Olivier Rouvière
- Hospices Civils de Lyon, Department of Urinary and Vascular Imaging, Hôpital Edouard Herriot, Lyon, France
| | - Ivo G Schoots
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Emma Jane Smith
- European Association of Urology Guidelines Office, Arnhem, The Netherlands
| | - Johan Stranne
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Urology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Catrin Tudur Smith
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | | | - Inge M Van Oort
- Radboud University Medical Center, Department of Urology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | - Cathy Y Yuan
- Department of Medicine, Health Science Centre, McMaster University, Hamilton, Ontario, Canada
| | | | - Ann M Henry
- University of Leeds, Leeds, UK; Leeds Cancer Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
17
|
Zheng X, Li C, Ai J, Dong G, Long M, Li M, Qiu S, Huang Y, Yang G, Zhang T, Li Z. No prognostic impact of staging bone scan in patients with stage IA non-small cell lung cancer. Ann Nucl Med 2024; 38:534-543. [PMID: 38602614 DOI: 10.1007/s12149-024-01927-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024]
Abstract
OBJECTIVE To investigate the survival benefit of preoperative bone scan in asymptomatic patients with early-stage non-small cell lung cancer (NSCLC). METHODS This retrospective study included patients with radical resection for stage T1N0M0 NSCLC between March 2013 and December 2018. During postoperative follow-up, we monitored patient survival and the development of bone metastasis. We compared overall survival, bone metastasis-free survival, and recurrence-free survival in patients with or without preoperative bone scan. Propensity score matching and inverse probability of treatment weighting were used to minimize election bias. RESULTS A total of 868 patients (58.19 ± 9.69 years; 415 men) were included in the study. Of 87.7% (761 of 868) underwent preoperative bone scan. In the multivariable analyses, bone scan did not improve overall survival (hazard ratio [HR] 1.49; 95% confidence intervals [CI] 0.91-2.42; p = 0.113), bone metastasis-free survival (HR 1.18; 95% CI 0.73-1.90; p = 0.551), and recurrence-free survival (HR 0.89; 95% CI 0.58-1.39; p = 0.618). Similar results were obtained after propensity score matching (overall survival [HR 1.28; 95% CI 0.74-2.23; p = 0.379], bone metastasis-free survival [HR 1.00; 95% CI 0.58-1.72; p = 0.997], and recurrence-free survival [HR 0.76; 95% CI 0.46-1.24; p = 0.270]) and inverse probability of treatment weighting. CONCLUSION There were no significant differences in overall survival, bone metastasis-free survival, and recurrence-free survival between asymptomatic patients with clinical stage IA NSCLC with or without preoperative bone scan.
Collapse
Affiliation(s)
- Xia Zheng
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China
| | - Chunxia Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Ai
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China
| | - Guili Dong
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China
| | - Man Long
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China
| | - Mingyi Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China
| | - Shilin Qiu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China
| | - Yanni Huang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Guangjun Yang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan, China.
| |
Collapse
|
18
|
Sutera P, Deek MP, Deek RA, Guler OC, Hurmuz P, Reyhan M, Rowe S, Radwan N, Dipasquale S, Hrinivich WT, Lowe K, Ren L, Saraiya B, Ennis R, Hathout L, Mayer T, Deweese TL, Song DY, Kiess A, Oymak E, Pienta K, Feng F, Pomper M, Ozyigit G, Tran PT, Onal C, Phillips RM. Prostate-Specific Membrane Antigen PET Response Associates with Metastasis-Free Survival After Stereotactic Ablative Radiation in Oligometastatic Prostate Cancer. Adv Radiat Oncol 2024; 9:101507. [PMID: 38799104 PMCID: PMC11127093 DOI: 10.1016/j.adro.2024.101507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/28/2024] [Indexed: 05/29/2024] Open
Abstract
Purpose Emerging data suggest that metastasis-directed therapy (MDT) improves outcomes in patients with oligometastatic castration-sensitive prostate cancer (omCSPC). Prostate-specific membrane antigen positron emission tomography (PSMA-PET) can detect occult metastatic disease, and PSMA response has been proposed as a biomarker for treatment response. Herein, we identify and validate a PSMA-PET biomarker for metastasis-free survival (MFS) following MDT in omCSPC. Methods and Materials We performed an international multi-institutional retrospective study of patients with omCSPC, defined as ≤3 lesions, treated with metastasis-directed stereotactic ablative radiation who underwent PSMA-PET/computed tomography (CT) before and after (median, 6.2 months; range, 2.4-10.9 months) treatment. Pre- and post-MDT PSMA-PET/CT maximum standardized uptake value (SUVmax) was measured for all lesions, and PSMA response was defined as the percent change in SUVmax of the least responsive lesion. PSMA response was both evaluated as a continuous variable and dichotomized into PSMA responders, with a complete/partial response (at least a 30% reduction in SUVmax), and PSMA nonresponders, with stable/progressive disease (less than a 30% reduction in SUVmax). PSMA response was correlated with conventional imaging-defined metastasis-free survival (MFS) via Kaplan-Meier and Cox regression analysis. Results A total of 131 patients with 261 treated metastases were included in the analysis, with a median follow-up of 29 months (IQR, 18.5-41.3 months). After stereotactic ablative radiation, 70.2% of patients were classified as PSMA responders. Multivariable analysis demonstrated that PSMA response as a continuous variable was associated with a significantly worse MFS (hazard ratio = 1.003; 95% CI, 1.001-1.006; P = .016). Patients classified as PSMA responders were found to have a significantly improved median MFS of 39.9 versus 12 months (P = .001) compared with PSMA nonresponders. Our study is limited as it is a retrospective review of a heterogenous population. Conclusions After stereotactic ablative radiation, PSMA-PET response appears to be a radiographic biomarker that correlates with MFS in omCSPC. This approach holds promise for guiding clinical management of omCSPC and should be validated in a prospective setting.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Matthew P. Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Rebecca A. Deek
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ozan Cem Guler
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana Dr Turgut Noyan Research and Treatment Center, Adana, Turkey
| | - Pervin Hurmuz
- Department of Radiation Oncology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Mehmet Reyhan
- Department of Nuclear Medicine, Faculty of Medicine, Baskent University, Adana Dr Turgut Noyan Research and Treatment Center, Adana, Turkey
| | - Steven Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Noura Radwan
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shirl Dipasquale
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William T. Hrinivich
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kathryn Lowe
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lei Ren
- Department of Radiation Oncology, University of Maryland, Baltimore, Maryland
| | - Biren Saraiya
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Ronald Ennis
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Lara Hathout
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Tina Mayer
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Theodore L. Deweese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel Y. Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ana Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ezgi Oymak
- Division of Radiation Oncology, Iskenderun Gelisim Hospital, Hatay, Turkey
| | - Kenneth Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Felix Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Martin Pomper
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gokhan Ozyigit
- Department of Radiation Oncology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Phuoc T. Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Radiation Oncology, University of Maryland, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cem Onal
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana Dr Turgut Noyan Research and Treatment Center, Adana, Turkey
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Ankara, Turkey
| | - Ryan M. Phillips
- Department of Radiation Oncology, The Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
19
|
Kwak L, Ravi P, Armstrong JG, Beckendorf V, Chin JL, D'Amico AV, Dearnaley DP, Di Stasi SM, Gillessen S, Lukka H, Mottet N, Pommier P, Seiferheld W, Sydes MR, Tombal B, Zapatero A, Regan MM, Xie W, Sweeney CJ. Prognostic Impact of Prostate-Specific Antigen at 6 Months After Radiotherapy in Localized Prostate Cancer: An Individual Patient Data Analysis of Randomized Trials. J Clin Oncol 2024; 42:2132-2138. [PMID: 38471051 DOI: 10.1200/jco.23.00762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 10/30/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024] Open
Abstract
PURPOSE We sought to evaluate the prognostic impact of prostate-specific antigen (PSA) at 6 months after completion of radiotherapy (RT) in patients treated with RT alone, RT plus short-term (st; 3-6 months), and RT plus long-term (lt; 24-36 months) androgen-deprivation therapy (ADT). PATIENTS AND METHODS Individual patient data were obtained from 16 randomized trials evaluating RT ± ADT for localized prostate cancer (PCa) between 1987 and 2011. The lowest PSA recorded within 6 months after RT completion was identified and categorized as < or ≥0.1 ng/mL. The primary outcomes were metastasis-free survival (MFS), PCa-specific mortality (PCSM), and overall survival (OS), from 12 months after random assignment. RESULTS Ninety-eight percent (n = 2,339/2,376) of patients allocated to RT alone, 84% (n = 4,756/5,658) allocated to RT + stADT, and 77% (n = 1,258/1,626) allocated to RT + ltADT had PSA ≥0.1 ng/mL within 6 months after completing RT. PSA ≥0.1 ng/mL was associated with lower MFS and OS and higher PCSM among patients allocated to RT ± ADT (RT - MFS: hazard ratio [HR], 2.24 [95% CI, 1.21 to 4.16]; PCSM: subdistribution hazard ratio [sHR], 1.82 [0.51 to 6.49]; OS: HR, 1.72 [0.97 to 3.05]; RT + stADT - MFS: HR, 1.27 [1.12 to 1.44]; PCSM: sHR, 2.10 [1.52 to 2.92]; OS: HR, 1.26 [1.11 to 1.44]; RT + ltADT - MFS: HR, 1.58 [1.27 to 1.96]; PCSM: sHR, 1.97 [1.11 to 3.49]; OS: HR, 1.59 [1.27 to 1.99]). Five-year MFS rates among patients allocated to RT, RT + stADT, and RT + ltADT were 91% versus 79%, 83% versus 76%, and 87% versus 74%, respectively, based on PSA < or ≥0.1 ng/mL. CONCLUSION PSA ≥0.1 ng/mL within 6 months after RT completion was prognostic for lt outcomes in patients treated with RT ± ADT for localized PCa. This can be used to counsel patients treated with RT ± ADT and in guiding clinical trial design evaluating novel systemic therapies with RT + ADT as well as (de)intensification strategies.
Collapse
Affiliation(s)
- Lucia Kwak
- Dana-Farber Cancer Institute, Boston, MA
| | | | - John G Armstrong
- Radiation Oncology Department, Cancer Trials Ireland, St Luke's Hospital, Dublin, Ireland
| | | | | | | | - David P Dearnaley
- The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | - Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Himanshu Lukka
- McMaster University and Juravinski Cancer Centre, Hamilton, ON, Canada
| | | | | | | | | | | | - Almudena Zapatero
- Department of Radiation Oncology, La Princesa University Hospital, Health Research Institute, Madrid, Spain
| | | | | | - Christopher J Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
20
|
Parker CC, Clarke NW, Cook AD, Kynaston H, Catton CN, Cross WR, Petersen PM, Persad RA, Saad F, Bower LC, Logue J, Payne H, Forcat S, Goldstein C, Murphy C, Anderson J, Barkati M, Bottomley DM, Branagan J, Choudhury A, Chung PWM, Cogley L, Goh CL, Hoskin P, Khoo V, Malone SC, Masters L, Morris SL, Nabid A, Ong AD, Raman R, Tarver KL, Tree AC, Worlding J, Wylie JP, Zarkar AM, Parulekar WR, Parmar MKB, Sydes MR. Adding 6 months of androgen deprivation therapy to postoperative radiotherapy for prostate cancer: a comparison of short-course versus no androgen deprivation therapy in the RADICALS-HD randomised controlled trial. Lancet 2024; 403:2405-2415. [PMID: 38763154 PMCID: PMC7616360 DOI: 10.1016/s0140-6736(24)00548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/07/2024] [Accepted: 03/15/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Previous evidence indicates that adjuvant, short-course androgen deprivation therapy (ADT) improves metastasis-free survival when given with primary radiotherapy for intermediate-risk and high-risk localised prostate cancer. However, the value of ADT with postoperative radiotherapy after radical prostatectomy is unclear. METHODS RADICALS-HD was an international randomised controlled trial to test the efficacy of ADT used in combination with postoperative radiotherapy for prostate cancer. Key eligibility criteria were indication for radiotherapy after radical prostatectomy for prostate cancer, prostate-specific antigen less than 5 ng/mL, absence of metastatic disease, and written consent. Participants were randomly assigned (1:1) to radiotherapy alone (no ADT) or radiotherapy with 6 months of ADT (short-course ADT), using monthly subcutaneous gonadotropin-releasing hormone analogue injections, daily oral bicalutamide monotherapy 150 mg, or monthly subcutaneous degarelix. Randomisation was done centrally through minimisation with a random element, stratified by Gleason score, positive margins, radiotherapy timing, planned radiotherapy schedule, and planned type of ADT, in a computerised system. The allocated treatment was not masked. The primary outcome measure was metastasis-free survival, defined as distant metastasis arising from prostate cancer or death from any cause. Standard survival analysis methods were used, accounting for randomisation stratification factors. The trial had 80% power with two-sided α of 5% to detect an absolute increase in 10-year metastasis-free survival from 80% to 86% (hazard ratio [HR] 0·67). Analyses followed the intention-to-treat principle. The trial is registered with the ISRCTN registry, ISRCTN40814031, and ClinicalTrials.gov, NCT00541047. FINDINGS Between Nov 22, 2007, and June 29, 2015, 1480 patients (median age 66 years [IQR 61-69]) were randomly assigned to receive no ADT (n=737) or short-course ADT (n=743) in addition to postoperative radiotherapy at 121 centres in Canada, Denmark, Ireland, and the UK. With a median follow-up of 9·0 years (IQR 7·1-10·1), metastasis-free survival events were reported for 268 participants (142 in the no ADT group and 126 in the short-course ADT group; HR 0·886 [95% CI 0·688-1·140], p=0·35). 10-year metastasis-free survival was 79·2% (95% CI 75·4-82·5) in the no ADT group and 80·4% (76·6-83·6) in the short-course ADT group. Toxicity of grade 3 or higher was reported for 121 (17%) of 737 participants in the no ADT group and 100 (14%) of 743 in the short-course ADT group (p=0·15), with no treatment-related deaths. INTERPRETATION Metastatic disease is uncommon following postoperative bed radiotherapy after radical prostatectomy. Adding 6 months of ADT to this radiotherapy did not improve metastasis-free survival compared with no ADT. These findings do not support the use of short-course ADT with postoperative radiotherapy in this patient population. FUNDING Cancer Research UK, UK Research and Innovation (formerly Medical Research Council), and Canadian Cancer Society.
Collapse
Affiliation(s)
- Chris C Parker
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - Noel W Clarke
- Department of Urology, The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK; Department of Urology, Salford Royal Hospital, Salford, UK
| | - Adrian D Cook
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Howard Kynaston
- Division of Cancer and Genetics, Cardiff University Medical School, Cardiff, UK
| | - Charles N Catton
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - William R Cross
- Department of Urology, St James's University Hospital, Leeds, UK
| | - Peter M Petersen
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Fred Saad
- Department of Urology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Lorna C Bower
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK; Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - John Logue
- Department of Urology, The Christie NHS Foundation Trust, Manchester, UK
| | | | - Silvia Forcat
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Cindy Goldstein
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Claire Murphy
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Juliette Anderson
- Department of Clinical Oncology, St James's University Hospital, Leeds, UK
| | - Maroie Barkati
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - David M Bottomley
- Department of Clinical Oncology, St James's University Hospital, Leeds, UK
| | | | - Ananya Choudhury
- Department of Urology, The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Peter W M Chung
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | | | | | - Peter Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Vincent Khoo
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - Shawn C Malone
- The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Lindsey Masters
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | | | - Abdenour Nabid
- Service de Radio-Oncologie, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Aldrich D Ong
- Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Rakesh Raman
- Kent Oncology Centre, Kent and Canterbury Hospital, Canterbury, UK
| | | | - Alison C Tree
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - Jane Worlding
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - James P Wylie
- Department of Urology, The Christie NHS Foundation Trust, Manchester, UK
| | - Anjali M Zarkar
- Department of Oncology, University Hospitals Birmingham, Birmingham, UK
| | - Wendy R Parulekar
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Mahesh K B Parmar
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK.
| |
Collapse
|
21
|
Pollack A, Pra AD. Androgen deprivation therapy combined with postoperative radiotherapy for prostate cancer management. Lancet 2024; 403:2353-2355. [PMID: 38763152 DOI: 10.1016/s0140-6736(24)00802-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 05/21/2024]
Affiliation(s)
- Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA.
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| |
Collapse
|
22
|
Correa RJM, Mendez LC. Helpful tool or blunt instrument?-the European Association of Urology Biochemical Recurrence Risk Classification as a decision-making tool for salvage radiotherapy. Transl Androl Urol 2024; 13:889-892. [PMID: 38855605 PMCID: PMC11157405 DOI: 10.21037/tau-23-665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/13/2024] [Indexed: 06/11/2024] Open
Affiliation(s)
- Rohann J M Correa
- Department of Radiation Oncology, London Health Sciences Centre, London, Canada
| | - Lucas C Mendez
- Department of Radiation Oncology, London Health Sciences Centre, London, Canada
| |
Collapse
|
23
|
Ravi P, Xie W, Buyse M, Halabi S, Kantoff PW, Sartor O, Attard G, Clarke N, D'Amico A, Dignam J, James N, Fizazi K, Gillessen S, Parulekar W, Sandler H, Spratt DE, Sydes MR, Tombal B, Williams S, Sweeney CJ. Refining Risk Stratification of High-risk and Locoregional Prostate Cancer: A Pooled Analysis of Randomized Trials. Eur Urol 2024:S0302-2838(24)02380-7. [PMID: 38777647 DOI: 10.1016/j.eururo.2024.04.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND OBJECTIVE Radiotherapy (RT) and long-term androgen deprivation therapy (ltADT; 18-36 mo) is a standard of care in the treatment of high-risk localized/locoregional prostate cancer (HRLPC). We evaluated the outcomes in patients treated with RT + ltADT to identify which patients have poorer prognosis with standard therapy. METHODS Individual patient data from patients with HRLPC (as defined by any of the following three risk factors [RFs] in the context of cN0 disease-Gleason score ≥8, cT3-4, and prostate-specific antigen [PSA] >20 ng/ml, or cN1 disease) treated with RT and ltADT in randomized controlled trials collated by the Intermediate Clinical Endpoints in Cancer of the Prostate group. The outcome measures of interest were metastasis-free survival (MFS), overall survival (OS), time to metastasis, and prostate cancer-specific mortality. Multivariable Cox and Fine-Gray regression estimated hazard ratios (HRs) for the three RFs and cN1 disease. KEY FINDINGS AND LIMITATIONS A total of 3604 patients from ten trials were evaluated, with a median PSA value of 24 ng/ml. Gleason score ≥8 (MFS HR = 1.45; OS HR = 1.42), cN1 disease (MFS HR = 1.86; OS HR = 1.77), cT3-4 disease (MFS HR = 1.28; OS HR = 1.22), and PSA >20 ng/ml (MFS HR = 1.30; OS HR = 1.21) were associated with poorer outcomes. Adjusted 5-yr MFS rates were 83% and 78%, and 10-yr MFS rates were 63% and 53% for patients with one and two to three RFs, respectively; corresponding 10-yr adjusted OS rates were 67% and 60%, respectively. In cN1 patients, adjusted 5- and 10-yr MFS rates were 67% and 36%, respectively, and 10-yr OS was 47%. CONCLUSIONS AND CLINICAL IMPLICATIONS HRLPC patients with two to three RFs (and cN0) or cN1 disease had the poorest outcomes on RT and ltADT. This will help in counseling patients treated in routine practice and in guiding adjuvant trials in HRLPC. PATIENT SUMMARY Radiotherapy and long-term hormone therapy are standard treatments for high-risk and locoregional prostate cancer. In this report, we defined prognostic groups within high-risk/locoregional prostate cancer and showed that outcomes to standard therapy are poorest in those with two or more "high-risk" factors or evidence of lymph node involvement. Such patients may therefore be the best candidates for intensification of treatment.
Collapse
Affiliation(s)
- Praful Ravi
- Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Wanling Xie
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marc Buyse
- International Drug Development Institute, Louvain-la-Neuve, Belgium; I-BioStat, Hasselt University, Hasselt, Belgium
| | | | - Philip W Kantoff
- Convergent Therapeutics, Cambridge, MA, USA; Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Noel Clarke
- The Christie NHS Foundation Trust, Manchester, UK
| | - Anthony D'Amico
- Dana-Farber Cancer Institute, Boston, MA, USA; Brigham & Women's Hospital, Boston, MA, USA
| | | | - Nicholas James
- The Institute of Cancer Research & The Royal Marsden NHS Foundation Trust, London, UK
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland
| | | | | | - Daniel E Spratt
- University Hospitals Siedman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | | | | | - Christopher J Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
24
|
Wang L. Instant Oncology: FLAME. Clin Oncol (R Coll Radiol) 2024; 36:271-272. [PMID: 38368228 DOI: 10.1016/j.clon.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 02/19/2024]
Affiliation(s)
- L Wang
- The Royal Marsden Hospital, Sutton, UK.
| |
Collapse
|
25
|
Jacques J, Sahki N, Meknaci É, Eschwege P, Peiffert D, Demogeot N. Brachytherapy for favorable prognostic prostate cancer in men up to 60 years of age: Long term follow-up. Brachytherapy 2024; 23:301-308. [PMID: 38480107 DOI: 10.1016/j.brachy.2024.01.010] [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: 07/07/2023] [Revised: 12/29/2023] [Accepted: 01/18/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE Brachytherapy (BT) is a standard treatment for low- and favorable intermediate-risk prostate adenocarcinoma. Few studies have focused on young patients. We therefore evaluated long-term efficacy and toxicity of BT in patients aged ≤ 60 years with low- and favorable intermediate-risk prostate cancer. MATERIALS AND METHODS This retrospective study included patients aged ≤60 years with low- or favorable intermediate-risk prostate adenocarcinoma treated with iodine BT alone between 1999 and 2014 at the Institut de Cancérologie de Lorraine. Follow-up assessment included incidence of biochemical failure (BF) at 10 and 15 years after BT, as well as survival data and late toxicities. RESULTS A total of 177 patients of median age 56 years (54-58) were analyzed, with a median follow-up of 126 months (97-172). Incidence of BF at 10 and 15 years after BT was 5.4% and 11.7% respectively. PSA nadir (HR = 51.8 [95% CI 6.69-277], p < 0.001), age at treatment (HR = 1.78 [95% CI 1.19-2.65], p = 0.005) and prostate D90% (HR = 1.08 [95% CI 1.01-1.15], p < 0.021) were identified as predictive factors of BF. Overall survival at 10 and 15 years after BT was 92.8% and 84.4% respectively. Cancer-specific survival at 10 and 15 years after BT was 99.3% and 97.7% respectively. No major toxicity was recorded. CONCLUSIONS Exclusive BT is a long-term effective treatment for patients aged ≤ 60 years with low- or favorable intermediate-risk prostate adenocarcinoma.
Collapse
Affiliation(s)
- Juliette Jacques
- Department of Radiotherapy, Institut de Cancérologie de Lorraine, Vandoeuvre-lès- Nancy, France; Université de Lorraine, Faculté de Médecine de Nancy, Vandoeuvre-lès-Nancy, France.
| | - Nassim Sahki
- Methodology Biostatistic unit, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Émilie Meknaci
- Department of Radiotherapy, Institut de Cancérologie de Lorraine, Vandoeuvre-lès- Nancy, France
| | - Pascal Eschwege
- Department of Radiotherapy, Institut de Cancérologie de Lorraine, Vandoeuvre-lès- Nancy, France; Université de Lorraine, Faculté de Médecine de Nancy, Vandoeuvre-lès-Nancy, France
| | - Didier Peiffert
- Department of Radiotherapy, Institut de Cancérologie de Lorraine, Vandoeuvre-lès- Nancy, France; Université de Lorraine, Faculté de Médecine de Nancy, Vandoeuvre-lès-Nancy, France; APEMAC, Université de Lorraine, Faculté de Médecine de Nancy, Vandoeuvre-lès-Nancy, France
| | - Nicolas Demogeot
- Department of Radiotherapy, Institut de Cancérologie de Lorraine, Vandoeuvre-lès- Nancy, France
| |
Collapse
|
26
|
Tward JD, Lenz L, Gutin A, Clegg W, Kasten CR, Finch R, Cohen T, Michalski J, Kishan AU. Using the Cell-Cycle Risk Score to Predict the Benefit of Androgen-Deprivation Therapy Added to Radiation Therapy in Patients With Newly Diagnosed Prostate Cancer. JCO Precis Oncol 2024; 8:e2300722. [PMID: 38748970 PMCID: PMC11371120 DOI: 10.1200/po.23.00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 09/05/2024] Open
Abstract
PURPOSE Guidelines recommend adding androgen-deprivation therapy (ADT) to radiation therapy (RT) in certain patients with localized prostate cancer. Individualized genomic testing may improve the prognostic accuracy of risk assessments. Herein, we describe a mathematical model of the benefit of adding ADT to RT as a function of the personalized clinical cell-cycle risk (CCR) score to inform 10-year metastasis risk. METHODS A model of absolute risk reduction (ARR) was built using a retrospective cohort of men tested with Prolaris who received RT alone (N = 467). The relative benefit of ADT added to RT to reduce distant metastasis was estimated at 41% on the basis of a meta-analysis of randomized trials. The ARR and number needed to treat (NNT) were computationally derived in patients clinically tested with Prolaris between January 1, 2020, and October 31, 2022 (N = 56,485). Risks were predicted using a cause-specific Cox proportional hazards model with CCR score predicting time to metastasis. A CCR score of 2.112 represents the validated multimodal treatment (MMT) threshold. RESULTS The ARR from ADT increased from almost zero at low CCR scores to 17.1% at CCR = 3.690 with the corresponding NNT = 6, indicating that adding ADT to RT would prevent metastasis within 10 years for one of every six treated individuals. In the clinical cohort, the average ARR was 0.86% in individuals under the MMT threshold (NNT = 116). The average ARR was 8.19% in individuals above the MMT threshold (NNT = 12). Broad ranges of ADT benefit were observed within National Comprehensive Cancer Network risk categories. CONCLUSION The precise and personalized risk estimate of metastasis provided by the CCR score can help inform patients and physicians when considering treatment intensification.
Collapse
|
27
|
James ND, Tannock I, N'Dow J, Feng F, Gillessen S, Ali SA, Trujillo B, Al-Lazikani B, Attard G, Bray F, Compérat E, Eeles R, Fatiregun O, Grist E, Halabi S, Haran Á, Herchenhorn D, Hofman MS, Jalloh M, Loeb S, MacNair A, Mahal B, Mendes L, Moghul M, Moore C, Morgans A, Morris M, Murphy D, Murthy V, Nguyen PL, Padhani A, Parker C, Rush H, Sculpher M, Soule H, Sydes MR, Tilki D, Tunariu N, Villanti P, Xie LP. The Lancet Commission on prostate cancer: planning for the surge in cases. Lancet 2024; 403:1683-1722. [PMID: 38583453 DOI: 10.1016/s0140-6736(24)00651-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/28/2023] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Affiliation(s)
- Nicholas D James
- Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK.
| | - Ian Tannock
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Felix Feng
- University of California, San Francisco, USA
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Syed Adnan Ali
- University of Manchester, Manchester, UK; The Christie Hospital, Manchester, UK
| | | | | | | | - Freddie Bray
- International Agency for Research on Cancer, Lyon, France
| | - Eva Compérat
- Tenon Hospital, Sorbonne University, Paris; AKH Medical University, Vienna, Austria
| | - Ros Eeles
- Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | | | | | | | - Áine Haran
- The Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | | | | | | | - Stacy Loeb
- New York University, New York, NY, USA; Manhattan Veterans Affairs, New York, NY, USA
| | | | | | | | - Masood Moghul
- Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | | | | | - Michael Morris
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Declan Murphy
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | | | | | | | | | | | | | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | | | - Derya Tilki
- Martini-Klinik Prostate Cancer Center and Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Türkiye
| | - Nina Tunariu
- Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | | | - Li-Ping Xie
- First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Lu Y, Jiang J, Yang G, Ding H, Zheng Q, Ji L, Wang Y, Dong Z, Zhai Z, Tian J, Zhang Y, Wang J, Yang L, Wang Z. Comparative effectiveness of multiple androgen receptor signaling inhibitor medicines with androgen deprivation therapy for metastatic hormone-sensitive prostate cancer: a study in the real world. Front Oncol 2024; 14:1324181. [PMID: 38699643 PMCID: PMC11063352 DOI: 10.3389/fonc.2024.1324181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/11/2024] [Indexed: 05/05/2024] Open
Abstract
Background The current treatment strategy for metastatic Hormone-Sensitive Prostate Cancer (mHSPC) is the combination of Androgen Receptor Signaling Inhibitors (ARSIs) medicines with androgen deprivation therapy (ADT). However, there is a lack of real-world data comparing the efficacy of different ARSI pharmaceuticals. Therefore, the objective of this study was to compare the effectiveness and safety of bicalutamide, abiraterone, enzalutamide, and apalutamide in combination with ADT for patients with mHSPC. Methods We retrospectively analyzed 82 patients diagnosed with mHSPC, including 18 patients treated with abiraterone acetate with prednisone, 21 patients with enzalutamide, 20 patients with apalutamide, and 23 patients with bicalutamide. We evaluated PSA progression-free survival (PSA-PFS), imaging progression-free survival (r PFS), castration resistance progression-free survival (CRPC-PFS), and overall survival (OS) using Kaplan-Meier survival analyses. Additionally, we explored relevant factors affecting prognosis through univariate and multivariate Cox risk-proportionality models. PSA response rates at 3, 6, and 12 months, nadir PSA levels (nPSA), and time to nadir (TTN) in different medication subgroups after treatment were documented, and we used one-way ANOVA to determine the effect of these measures on patient prognosis. Results In comparison with bicalutamide, both enzalutamide and apalutamide have shown significant advantages in delaying disease progression among mHSPC patients. Specifically, enzalutamide has been found to significantly prolong PSA-PFS (HR 2.244; 95% CI 1.366-3.685, p=0.001), rPFS (HR 2.539; 95% CI 1.181-5.461; p= 0.007), CRPC-PFS (HR 2.131; 95% CI 1.295-3.506; p= 0.003), and OS (HR 2.06; 95% CI 1.183-3.585; P=0.005). Similarly, apalutamide has significantly extended PSA-PFS (HR 5.071; 95% CI 1.711-15.032; P= 0.003) and CRPC-PFS (HR 6.724; 95% CI 1.976-22.878; P=0.002) among patients. On the other hand, the use of abiraterone in combination with ADT did not demonstrate a significant advantage in delaying diseases progression when compared with the other three agents in mHSPC patients. There were no significant differences in overall adverse event rates among the four pharmaceuticals in terms of safety. Additionally, the observation of PSA kinetics revealed that enzalutamide, apalutamide, and abiraterone acetate had a significant advantage in achieving deep PSA response (PSA ≤ 0.2 ng/ml) compared with bicalutamide (p=0.007 at 12 months). Enzalutamide and apalutamide exhibited preeminence efficacy, with no substantial difference observed between the two medications. Conclusions Abiraterone, enzalutamide, and apalutamide were found to significantly reduce and stabilize PSA levels in mHSPC patients more quickly and thoroughly than bicalutamide. Furthermore, enzalutamide and apalutamide were found to significantly prolong survival and delay disease progression in mHSPC patients compared with bicalutamide. It should be noted that abiraterone did not demonstrate a significant advantage in delaying disease compared with enzalutamide and apalutamide. After conducting drug toxicity analyses, it was determined that there were no significant differences among the four drugs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Zhiping Wang
- Department of Urology, Gansu Provincial Key Laboratory of Urological Disease Research, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
29
|
Aggarwal R, Heller G, Hillman DW, Xiao H, Picus J, Taplin ME, Dorff T, Appleman L, Weckstein D, Patnaik A, Bryce A, Shevrin D, Mohler J, Anderson D, Rao A, Tagawa S, Tan A, Halabi S, Dooley K, O'Brien P, Chen R, Ryan CJ, Eggener SE, Morris MJ. PRESTO: A Phase III, Open-Label Study of Intensification of Androgen Blockade in Patients With High-Risk Biochemically Relapsed Castration-Sensitive Prostate Cancer (AFT-19). J Clin Oncol 2024; 42:1114-1123. [PMID: 38261983 DOI: 10.1200/jco.23.01157] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/01/2023] [Accepted: 11/08/2023] [Indexed: 01/25/2024] Open
Abstract
PURPOSE Patients with biochemically recurrent prostate cancer (BRPC) after radical prostatectomy and a short PSA doubling time are at risk for distant metastases. Apalutamide, an androgen receptor antagonist, and abiraterone acetate plus prednisone (AAP) prolong survival in the metastatic setting. We evaluated whether intensification of androgen-deprivation therapy (ADT) improves outcomes in BRPC. PATIENTS AND METHODS PRESTO is a randomized phase III, open-label trial in patients with BRPC and PSA doubling time ≤9 months (ClinicalTrials.gov identifier: NCT03009981). Patients were randomly assigned 1:1:1 to receive a finite 52-week treatment course with ADT control, ADT + apalutamide, or ADT + apalutamide + AAP. The primary end point was PSA progression-free survival (PSA-PFS), defined as serum PSA >0.2 ng/mL after treatment completion. RESULTS Five hundred three patients were enrolled. The median PSA was 1.8 ng/mL (IQR, 1.0-3.6). At the first planned interim analysis, both experimental arms significantly prolonged PSA-PFS compared with the control arm (median, 24.9 months for ADT + apalutamide v 20.3 months for ADT; hazard ratio [HR], 0.52 [95% CI, 0.35 to 0.77]; P = .00047; median, 26.0 months for ADT + apalutamide + AAP v 20.0 months for ADT; HR, 0.48 [95% CI, 0.32 to 0.71]; P = .00008). Median time to testosterone recovery did not differ across treatment arms. The most common grade ≥3 adverse event was hypertension (7.5%, 7.4%, and 18% in ADT, ADT + apalutamide, and ADT + apalutamide + AAP arms, respectively). CONCLUSION Intensified AR blockade for a finite duration prolongs PSA-PFS with a manageable safety profile, without adversely affecting time to testosterone recovery. The addition of apalutamide to ADT should be considered in patients with high-risk BRPC.
Collapse
Affiliation(s)
| | - Glenn Heller
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Han Xiao
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | | | | | | | | | - Arpit Rao
- Baylor College of Medicine, Houston, TX
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Blondeaux E, Xie W, Carmisciano L, Mura S, Sanna V, De Laurentiis M, Caputo R, Turletti A, Durando A, De Placido S, De Angelis C, Bisagni G, Gasparini E, Rimanti A, Puglisi F, Mansutti M, Landucci E, Fabi A, Arecco L, Perachino M, Bruzzone M, Boni L, Lambertini M, Del Mastro L, Regan MM. Intermediate clinical endpoints in early-stage breast cancer: an analysis of individual patient data from the Gruppo Italiano Mammella and Mammella Intergruppo trials. EClinicalMedicine 2024; 70:102501. [PMID: 38685923 PMCID: PMC11056413 DOI: 10.1016/j.eclinm.2024.102501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 05/02/2024] Open
Abstract
Background Intermediate clinical endpoints (ICEs) are frequently used as primary endpoint in randomised trials (RCTs). We aim to assess whether changes in different ICEs can be used to predict changes in overall survival (OS) in adjuvant breast cancer trials. Methods Individual patient level data from adjuvant phase III RCTs conducted by the Gruppo Italiano Mammella (GIM) and Mammella Intergruppo (MIG) study groups were used. ICEs were computed according to STEEP criteria. Using a two-stage meta-analytic model, we assessed the surrogacy of each ICE at both the outcome (i.e., OS and ICE are correlated irrespective of treatment) and trial (i.e., treatment effects on ICE and treatment effect on OS are correlated) levels. The following ICEs were considered as potential surrogate endpoints of OS: disease-free survival (DFS), distant disease-free survival (DDFS), distant relapse-free survival (DRFS), recurrence-free survival (RFS), recurrence-free interval (RFI), distant recurrence-free interval (DRFI), breast cancer-free interval (BCFI), and invasive breast cancer-free survival (IBCFS). The estimates of the degree of correlation were obtained by copula models and weighted linear regression. Kendall's τ and R2 ≥ 0.70 were considered as indicators of a clinically relevant surrogacy. Findings Among the 12,397 patients enrolled from November 1992 to July 2012 in six RCTs, median age at enrolment was 57 years (interquartile range (IQR) 49-65). After a median follow-up of 10.3 years (IQR 6.4-14.5), 2131 (17.2%) OS events were observed, with 1390 (65.2%) attributed to breast cancer. At the outcome-level, Kendall's τ ranged from 0.69 for BCFI to 0.84 for DRFS. For DFS, DDFS, DRFS, RFS, RFI, DRFI, BCFI, and IBCFS endpoints, over 95% of the 8-year OS variability was attributable to the variation of the 5-year ICE. At the trial-level, treatment effects for the different ICEs and OS were strongly correlated, with the highest correlation for RFS and DRFS and the lowest for BCFI. Interpretation Our results provide evidence supporting the use of DFS, DDFS, DRFS, RFS, RFI, DRFI, and IBCFS as primary endpoint in breast cancer adjuvant trials. Funding This analysis was supported by the Italian Association for Cancer Research ("Associazione Italiana per la Ricerca sul Cancro", AIRC; IG 2017/20760) and by Italian Ministry of Health-5 × 1000 funds (years 2021-2022).
Collapse
Affiliation(s)
- Eva Blondeaux
- U.O. Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Wanling Xie
- Division of Biostatistics, Dana-Farber Cancer Institute, Boston, USA
| | - Luca Carmisciano
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Silvia Mura
- Department of Medical Oncology, UOC Oncologia Medica, University Hospital of Sassari, Sassari, Italy
| | - Valeria Sanna
- Department of Medical Oncology, UOC Oncologia Medica, University Hospital of Sassari, Sassari, Italy
| | - Michelino De Laurentiis
- Istituto Nazionale per lo Studio e la Cura dei Tumouri, Fondazione Pascale IRCCS, Napoli, Italy
| | - Roberta Caputo
- Istituto Nazionale per lo Studio e la Cura dei Tumouri, Fondazione Pascale IRCCS, Napoli, Italy
| | - Anna Turletti
- Medical Oncology, Ospedale Martini ASL Città di Torino, Torino, Italy
| | - Antonio Durando
- Breast Unit, Città della Salute e della Scienza, Ospedale S. Anna, Torino, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Giancarlo Bisagni
- Department of Oncology and Advanced Technology, Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisa Gasparini
- Department of Oncology and Advanced Technology, Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Anita Rimanti
- ASST Mantova, Azienda Ospedaliera Carlo Poma, Mantova, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, Unit of Medical Oncology and Cancer Prevention, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Mauro Mansutti
- Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | | | - Alessandra Fabi
- Precision Medicine Breast Unit, Scientific Directorate, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Arecco
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Marta Perachino
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Marco Bruzzone
- U.O. Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Boni
- U.O. Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Meredith M. Regan
- Division of Biostatistics, Dana-Farber Cancer Institute, Boston, USA
| |
Collapse
|
31
|
Halabi S, Roy A, Rydzewska L, Guo S, Godolphin P, Hussain M, Tangen C, Thompson I, Xie W, Carducci MA, Smith MR, Morris MJ, Gravis G, Dearnaley DP, Verhagen P, Goto T, James N, Buyse ME, Tierney JF, Sweeney C. Radiographic Progression-Free Survival and Clinical Progression-Free Survival as Potential Surrogates for Overall Survival in Men With Metastatic Hormone-Sensitive Prostate Cancer. J Clin Oncol 2024; 42:1044-1054. [PMID: 38181323 PMCID: PMC10950170 DOI: 10.1200/jco.23.01535] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/27/2023] [Accepted: 10/18/2023] [Indexed: 01/07/2024] Open
Abstract
PURPOSE Despite major increases in the longevity of men with metastatic hormone-sensitive prostate cancer (mHSPC), most men still die of prostate cancer. Phase III trials assessing new therapies in mHSPC with overall survival (OS) as the primary end point will take approximately a decade to complete. We investigated whether radiographic progression-free survival (rPFS) and clinical PFS (cPFS) are valid surrogates for OS in men with mHSPC and could potentially be used to expedite future phase III clinical trials. METHODS We obtained individual patient data (IPD) from 9 eligible randomized trials comparing treatment regimens (different androgen deprivation therapy [ADT] strategies or ADT plus docetaxel in the control or research arms) in mHSPC. rPFS was defined as the time from random assignment to radiographic progression or death from any cause whichever occurred first; cPFS was defined as the time from random assignment to the date of radiographic progression, symptoms, initiation of new treatment, or death, whichever occurred first. We implemented a two-stage meta-analytic validation model where conditions of patient-level and trial-level surrogacy had to be met. We then computed the surrogate threshold effect (STE). RESULTS IPD from 6,390 patients randomly assigned from 1994 to 2012 from 13 units were pooled for a stratified analysis. The median OS, rPFS, and cPFS were 4.3 (95% CI, 4.2 to 4.5), 2.4 (95% CI, 2.3 to 2.5), and 2.3 years (95% CI, 2.2 to 2.4), respectively. The STEs were 0.80 and 0.81 for rPFS and cPFS end points, respectively. CONCLUSION Both rPFS and cPFS appear to be promising surrogate end points for OS. The STE of 0.80 or higher makes it viable for either rPFS or cPFS to be used as the primary end point that is surrogate for OS in phase III mHSPC trials with testosterone suppression alone as the backbone therapy and would expedite trial conduct.
Collapse
Affiliation(s)
- Susan Halabi
- Duke University Medical Center, Duke University, Durham, NC
| | - Akash Roy
- Duke University Medical Center, Duke University, Durham, NC
| | - Larysa Rydzewska
- Medical Research Council Clinical Trials Unit at University College London, London, United Kingdom
| | - Siyuan Guo
- Duke University Medical Center, Duke University, Durham, NC
| | - Peter Godolphin
- Medical Research Council Clinical Trials Unit at University College London, London, United Kingdom
| | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | | | | | | | | | - Michael J. Morris
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gwenaelle Gravis
- Institut Paoli-Calmettes Aix-Mareseille Université, Marseille, France
| | - David P. Dearnaley
- Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | - Takayuki Goto
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nick James
- Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Marc E. Buyse
- International Drug Development Institute, Louvain-La-Neuve, Belgium
| | - Jayne F. Tierney
- Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | |
Collapse
|
32
|
Shore ND, Antonarakis ES, Ross AE, Marshall CH, Stratton KL, Ayanambakkam A, Cookson MS, McKay RR, Bryce AH, Kaymakcalan MD. A multidisciplinary approach to address unmet needs in the management of patients with non-metastatic castration-resistant prostate cancer. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00803-5. [PMID: 38431761 DOI: 10.1038/s41391-024-00803-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND With the availability of second-generation androgen receptor inhibitors (SGARIs), the treatment landscape has changed dramatically for patients with nonmetastatic castration-resistant prostate cancer (nmCRPC). In clinical trials, the SGARIs (apalutamide, enzalutamide, darolutamide) increased metastasis-free survival (MFS), overall survival (OS), and patient quality of life compared to placebo. These drugs were subsequently integrated into nmCRPC clinical practice guidelines. With advances in radiographic imaging, disease assessment, and patient monitoring, nmCRPC strategies are evolving to address limitations related to tracking disease progression using prostate-specific antigen (PSA) kinetics. METHODS A panel of 10 multidisciplinary experts in prostate cancer conducted reviews and discussions of unmet needs in the management and monitoring of patients with nmCRPC in order to develop consensus recommendations. RESULTS Across the SGARI literature, patient MFS and OS are generally comparable for all treatments, but important distinctions exist regarding short- and long-term drug safety profiles and drug-drug interactions. With respect to disease monitoring, a substantial proportion of patients using SGARIs may experience disease progression without rising PSA levels, suggesting a need for enhanced radiographic imaging in addition to PSA monitoring. Recent data also indicate that novel prostate-specific membrane antigen positron emission tomography radiotracers provide enhanced accuracy for disease detection, as compared to conventional imaging. CONCLUSIONS Clinical decision-making in nmCRPC has become more complex, with new opportunities to apply precision medicine to patient care. Multidisciplinary teams can ensure that patients with nmCRPC receive optimal and individualized disease management.
Collapse
Affiliation(s)
| | | | - Ashley E Ross
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Kelly L Stratton
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Michael S Cookson
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rana R McKay
- University of California, San Diego, La Jolla, CA, USA
| | | | | |
Collapse
|
33
|
Xie W, Ravi P, Buyse M, Halabi S, Kantoff P, Sartor O, Soule H, Clarke N, Dignam J, James N, Fizazi K, Gillessen S, Mottet N, Murphy L, Parulekar W, Sandler H, Tombal B, Williams S, Sweeney CJ. Validation of metastasis-free survival as a surrogate endpoint for overall survival in localized prostate cancer in the era of docetaxel for castration-resistant prostate cancer. Ann Oncol 2024; 35:285-292. [PMID: 38061427 PMCID: PMC10922430 DOI: 10.1016/j.annonc.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Prior work from the Intermediate Clinical Endpoints in Cancer of the Prostate (ICECaP) consortium (ICECaP-1) demonstrated that metastasis-free survival (MFS) is a valid surrogate for overall survival (OS) in localized prostate cancer (PCa). This was based on data from patients treated predominantly before 2004, prior to docetaxel being available for the treatment of metastatic castrate-resistant prostate cancer (mCRPC). We sought to validate surrogacy in a more contemporary era (ICECaP-2) with greater availability of docetaxel and other systemic therapies for mCRPC. PATIENTS AND METHODS Eligible trials for ICECaP-2 were those providing individual patient data (IPD) after publication of ICECaP-1 and evaluating adjuvant/salvage therapy for localized PCa, and which collected MFS and OS data. MFS was defined as distant metastases or death from any cause, and OS was defined as death from any cause. Surrogacy was evaluated using a meta-analytic two-stage validation model, with an R2 ≥ 0.7 defined a priori as clinically relevant. RESULTS A total of 15 164 IPD from 14 trials were included in ICECaP-2, with 70% of patients treated after 2004. The median follow-up was 8.3 years and the median postmetastasis survival was 3.1 years in ICECaP-2, compared with 1.9 years in ICECaP-1. For surrogacy condition 1, Kendall's tau was 0.92 for MFS with OS at the patient level, and R2 from weighted linear regression (WLR) of 8-year OS on 5-year MFS was 0.73 (95% confidence interval 0.53-0.82) at the trial level. For condition 2, R2 was 0.83 (95% confidence interval 0.64-0.89) from WLR of log[hazard ratio (HR)]-OS on log(HR)-MFS. The surrogate threshold effect on OS was an HR(MFS) of 0.81. CONCLUSIONS MFS remained a valid surrogate for OS in a more contemporary era, where patients had greater access to docetaxel and other systemic therapies for mCRPC. This supports the use of MFS as the primary outcome measure for ongoing adjuvant trials in localized PCa.
Collapse
Affiliation(s)
- W Xie
- Dana-Farber Cancer Institute, Boston, USA
| | - P Ravi
- Dana-Farber Cancer Institute, Boston, USA
| | - M Buyse
- International Drug Development Institute, Louvain-la-Neuve; I-BioStat, Hasselt University, Hasselt, Belgium
| | | | | | | | - H Soule
- Prostate Cancer Foundation, Santa Monica, USA
| | - N Clarke
- The Christie NHS Foundation Trust, Manchester, UK
| | - J Dignam
- University of Chicago, Chicago, USA
| | - N James
- The Institute of Cancer Research & The Royal Marsden NHS Foundation Trust, London, UK
| | - K Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - S Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona; Università della Svizzera Italiana, Lugano, Switzerland
| | - N Mottet
- Mutualite Francoise Loire, St Etienne, France
| | - L Murphy
- Medical Research Council at UCL, London, UK
| | - W Parulekar
- Queens University, Kingston, Ontario, Canada
| | - H Sandler
- Cedars-Sinai Medical Center, Los Angeles, USA
| | - B Tombal
- Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - S Williams
- Peter MacCallum Cancer Centre, Melbourne
| | - C J Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
34
|
Koerber SA, Höcht S, Aebersold D, Albrecht C, Boehmer D, Ganswindt U, Schmidt-Hegemann NS, Hölscher T, Mueller AC, Niehoff P, Peeken JC, Pinkawa M, Polat B, Spohn SKB, Wolf F, Zamboglou C, Zips D, Wiegel T. Prostate cancer and elective nodal radiation therapy for cN0 and pN0-a never ending story? : Recommendations from the prostate cancer expert panel of the German Society of Radiation Oncology (DEGRO). Strahlenther Onkol 2024; 200:181-187. [PMID: 38273135 PMCID: PMC10876748 DOI: 10.1007/s00066-023-02193-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024]
Abstract
For prostate cancer, the role of elective nodal irradiation (ENI) for cN0 or pN0 patients has been under discussion for years. Considering the recent publications of randomized controlled trials, the prostate cancer expert panel of the German Society of Radiation Oncology (DEGRO) aimed to discuss and summarize the current literature. Modern trials have been recently published for both treatment-naïve patients (POP-RT trial) and patients after surgery (SPPORT trial). Although there are more reliable data to date, we identified several limitations currently complicating the definitions of general recommendations. For patients with cN0 (conventional or PSMA-PET staging) undergoing definitive radiotherapy, only men with high-risk factors for nodal involvement (e.g., cT3a, GS ≥ 8, PSA ≥ 20 ng/ml) seem to benefit from ENI. For biochemical relapse in the postoperative situation (pN0) and no PSMA imaging, ENI may be added to patients with risk factors according to the SPPORT trial (e.g., GS ≥ 8; PSA > 0.7 ng/ml). If PSMA-PET/CT is negative, ENI may be offered for selected men with high-risk factors as an individual treatment approach.
Collapse
Affiliation(s)
- S A Koerber
- Department of Radiation Oncology, Barmherzige Brüder Hospital Regensburg, Prüfeninger Straße 86, 93049, Regensburg, Germany.
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| | - S Höcht
- Department of Radiation Oncology, Ernst von Bergmann Hospital Potsdam, Charlottenstraße 72, 14467, Potsdam, Germany
| | - D Aebersold
- Department of Radiation Oncology, Inselspital-Bern University Hospital, University of Bern, Freiburgstraße 4, 3010, Bern, Switzerland
| | - C Albrecht
- Nordstrahl Radiation Oncology Unit, Nürnberg North Hospital, Prof.-Ernst-Nathan-Str. 1, 90149, Nürnberg, Germany
| | - D Boehmer
- Department of Radiation Oncology, University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - U Ganswindt
- Department of Radiation Oncology, University Hospital Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - N-S Schmidt-Hegemann
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - T Hölscher
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Fiedlerstraße 19, 01307, Dresden, Germany
| | - A-C Mueller
- Department of Radiation Oncology, RKH Hospital Ludwigsburg, Posilipostraße 4, 71640, Ludwigsburg, Germany
| | - P Niehoff
- Department of Radiation Oncology, Sana Hospital Offenbach, Starkenburgring 66, 63069, Offenbach, Germany
| | - J C Peeken
- Department of Radiation Oncology, Technische Universität München, Ismaninger Straße 22, 81675, Munich, Germany
| | - M Pinkawa
- Department of Radiation Oncology, Robert Janker Klinik, Villenstraße 8, 53129, Bonn, Germany
| | - B Polat
- Department of Radiation Oncology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080, Würzburg, Germany
| | - S K B Spohn
- Department of Radiation Oncology, University Hospital Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
| | - F Wolf
- Department of Radiation Oncology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| | - C Zamboglou
- Department of Radiation Oncology, University Hospital Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Oncology Center, 1, Nikis Avenue, Agios Athanasios, 4108, Limassol, Cyprus
| | - D Zips
- Department of Radiation Oncology, University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - T Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| |
Collapse
|
35
|
Porreca A, Di Nicola M, Lucarelli G, Dorin VM, Soria F, Terracciano D, Mistretta FA, Luzzago S, Buonerba C, Cantiello F, Mari A, Minervini A, Veccia A, Antonelli A, Musi G, Hurle R, Busetto GM, Del Giudice F, Ferretti S, Perdonà S, Prete PD, Porreca A, Bove P, Crisan N, Russo GI, Damiano R, Amparore D, Porpiglia F, Autorino R, Piccinelli M, Brescia A, Tătaru SO, Crocetto F, Giudice AL, de Cobelli O, Schips L, Ferro M, Marchioni M. Time to progression is the main predictor of survival in patients with high-risk nonmuscle invasive bladder cancer: Results from a machine learning-based analysis of a large multi-institutional database. Urol Oncol 2024; 42:69.e17-69.e25. [PMID: 38302296 DOI: 10.1016/j.urolonc.2024.01.001] [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: 06/10/2023] [Revised: 11/13/2023] [Accepted: 01/01/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND In patients affected by high-risk nonmuscle invasive bladder cancer (HR-NMIBC) progression to muscle invasive status is considered as the main indicator of local treatment failure. We aimed to investigate the effect of progression and time to progression on overall survival (OS) and to investigate their validity as surrogate endpoints. METHODS A total of 1,510 patients from 18 different institutions treated for T1 high grade NMIBC, followed by a secondary transurethral resection and BCG intravesical instillation. We relied on random survival forest (RSF) to rank covariates based on OS prediction. Cox's regression models were used to quantify the effect of covariates on mortality. RESULTS During a median follow-up of 49.0 months, 485 (32.1%) patients progressed to MIBC, while 163 (10.8%) patients died. The median time to progression was 82 (95%CI: 78.0-93.0) months. In RSF time-to-progression and age were the most predictive covariates of OS. The survival tree defined 5 groups of risk. In multivariable Cox's regression models accounting for progression status as time-dependent covariate, shorter time to progression (as continuous covariate) was associated with longer OS (HR: 9.0, 95%CI: 3.0-6.7; P < 0.001). Virtually same results after time to progression stratification (time to progression ≥10.5 months as reference). CONCLUSION Time to progression is the main predictor of OS in patients with high risk NMIBC treated with BCG and might be considered a coprimary endpoint. In addition, models including time to progression could be considered for patients' stratification in clinical practice and at the time of clinical trials design.
Collapse
Affiliation(s)
- Annamaria Porreca
- Biostatistics Laboratory, Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Marta Di Nicola
- Biostatistics Laboratory, Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Vartolomei Mihai Dorin
- Department of Cell and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology from Târgu Mureș, Târgu Mureș, Romania
| | - Francesco Soria
- Division of Urology, Department of Surgical Sciences, Torino School of Medicine, Torino, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University Federico II, Napoli, Italy
| | | | - Stefano Luzzago
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Carlo Buonerba
- CRTR Rare Tumors Reference Center, AOU Federico II, Napoli, Italy
| | - Francesco Cantiello
- Department of Urology, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Andrea Mari
- Unit of Oncologic Minimally-Invasive Urology and Andrology, Department of Urology, Careggi Hospital, University of Florence, Florence, Italy
| | - Andrea Minervini
- Unit of Oncologic Minimally-Invasive Urology and Andrology, Department of Urology, Careggi Hospital, University of Florence, Florence, Italy
| | | | | | - Gennaro Musi
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Rodolfo Hurle
- Department of Urology, Humanitas Clinical and Research Hospital, Rozzano, Milan, Italy
| | | | | | - Simone Ferretti
- Urology Unit, Department of Medical, Oral and Biotechnological Sciences, "SS. Annunziata" Hospital, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Sisto Perdonà
- Uro-Gynecological Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Naples, Italy
| | - Paola Del Prete
- Scientific Directorate, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Naples, Italy
| | - Angelo Porreca
- Department of Robotic Urologic Surgery, Abano Terme Hospital, Abano Terme, Italy
| | - Pierluigi Bove
- Division of Urology, Department of Experimental Medicine and Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Nicolae Crisan
- Department of Urology, University of Medicine and Pharmacy Iuliu Haţieganu, Cluj-Napoca, Romania
| | | | - Rocco Damiano
- Department of Urology, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Daniele Amparore
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Hospital, University of Turin, Orbassano, Turin, Italy
| | - Francesco Porpiglia
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Hospital, University of Turin, Orbassano, Turin, Italy
| | | | - Mattia Piccinelli
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Antonio Brescia
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Sabin Octavian Tătaru
- I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Sciences and Technology from Târgu Mureș, Târgu Mureș, Romania
| | - Felice Crocetto
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy
| | | | - Ottavio de Cobelli
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Luigi Schips
- Urology Unit, Department of Medical, Oral and Biotechnological Sciences, "SS. Annunziata" Hospital, "G. d'Annunzio" University of Chieti, Chieti, Italy
| | - Matteo Ferro
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Michele Marchioni
- Urology Unit, Department of Medical, Oral and Biotechnological Sciences, "SS. Annunziata" Hospital, "G. d'Annunzio" University of Chieti, Chieti, Italy.
| |
Collapse
|
36
|
Krafft U, Hadaschik BA, Lückerath K, Herrmann K. A New Chapter in Neoadjuvant Therapy for High-risk Prostate Cancer? Eur Urol 2024; 85:227-228. [PMID: 37833179 DOI: 10.1016/j.eururo.2023.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023]
Affiliation(s)
- Ulrich Krafft
- Department of Urology, University Hospital Essen, University Duisburg-Essen, Essen, Germany; West German Cancer Center, University Dusiburg-Essen, University Hospital Essen, Germany.
| | - Boris A Hadaschik
- Department of Urology, University Hospital Essen, University Duisburg-Essen, Essen, Germany; West German Cancer Center, University Dusiburg-Essen, University Hospital Essen, Germany
| | - Katharina Lückerath
- West German Cancer Center, University Dusiburg-Essen, University Hospital Essen, Germany; Department of Nuclear Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- West German Cancer Center, University Dusiburg-Essen, University Hospital Essen, Germany; Department of Nuclear Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
37
|
Gharzai LA, Morris E, Suresh K, Nguyen-Tân PF, Rosenthal DI, Gillison ML, Harari PM, Garden AS, Koyfman S, Caudell JJ, Jones CU, Mitchell DL, Krempl G, Ridge JA, Gensheimer MF, Bonner JA, Filion E, Dunlap NE, Stokes WA, Le QT, Torres-Saavedra P, Mierzwa M, Schipper MJ. Surrogate endpoints in clinical trials of p16-positive squamous cell carcinoma of the oropharynx: an individual patient data meta-analysis. Lancet Oncol 2024; 25:366-375. [PMID: 38423050 PMCID: PMC10962533 DOI: 10.1016/s1470-2045(24)00016-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/19/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND The increased incidence of human papillomavirus (HPV)-related cancers has motivated efforts to optimise treatment for these patients with excellent prognosis. Validation of surrogates for overall survival could expedite the investigation of new therapies. We sought to evaluate candidate intermediate clinical endpoints in trials assessing definitive treatment of p16-positive oropharyngeal cancer with chemotherapy or radiotherapy. METHODS We did a retrospective review of five multicentre, randomised trials (NRG/RTOG 9003, 0129, 0234, 0522, and 1016) that tested radiotherapy with or without chemotherapy in patients (aged ≥18 years) with p16-positive localised head or neck squamous-cell carcinomas. Eight intermediate clinical endpoints were considered as potential surrogates for overall survival: freedom from local progression, freedom from regional progression, freedom from distant metastasis, freedom from locoregional progression, freedom from any progression, locoregional progression-free survival, progression-free survival, and distant metastasis-free survival. We used a two-stage meta-analytical framework, which requires high correlation between the intermediate clinical endpoint and overall survival at the patient level (condition 1), and high correlation between the treatment effect on the intermediate clinical endpoint and the treatment effect on overall survival (condition 2). For both, an r2 greater than 0·7 was used as criteria for clinically relevant surrogacy. FINDINGS We analysed 1373 patients with oropharyngeal cancer from May 9, 2020, to Nov 22, 2023. 1231 (90%) of patients were men, 142 (10%) were women, and 1207 (88%) were White, with a median age of 57 years (IQR 51-62). Median follow-up was 4·2 years (3·1-5·1). For the first condition, correlating the intermediate clinical endpoints with overall survival at the individual and trial level, the three composite endpoints of locoregional progression-free survival (Kendall's τ 0·91 and r2 0·72), distant metastasis-free survival (Kendall's τ 0·93 and r2 0·83), and progression-free survival (Kendall's τ 0·88 and r2 0·70) were highly correlated with overall survival at the patient level and at the trial-group level. For the second condition, correlating treatment effects of the intermediate clinical endpoints and overall survival, the composite endpoints of locoregional progression-free survival (r2 0·88), distant metastasis-free survival (r2 0·96), and progression-free survival (r2 0·92) remained strong surrogates. Treatment effects on the remaining intermediate clinical endpoints were less strongly correlated with overall survival. INTERPRETATION We identified locoregional progression-free survival, distant metastasis-free survival, and progression-free survival as surrogates for overall survival in p16-positive oropharyngeal cancers treated with chemotherapy or radiotherapy, which could serve as clinical trial endpoints. FUNDING NRG Oncology Operations, NRG Oncology SDMC, the National Cancer Institute, Eli Lilly, Aventis, and the University of Michigan.
Collapse
Affiliation(s)
- Laila A Gharzai
- Department of Radiation Oncology, Northwestern University, Chicago, IL, USA
| | - Emily Morris
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Krithika Suresh
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA; Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Phuc Felix Nguyen-Tân
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - David I Rosenthal
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maura L Gillison
- Department of Thoracic and Head/Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul M Harari
- Department of Radiation Oncology, University of Wisconsin, Madison, WI, USA
| | - Adam S Garden
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shlomo Koyfman
- Department of Radiation Oncology, University of Cleveland Medical Center, Cleveland, OH, USA
| | - Jimmy J Caudell
- Department of Radiation Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Christopher U Jones
- Department of Radiation Oncology, Sutter Cancer Research Consortium, Novato, CA, USA
| | - Darrion L Mitchell
- Department of Radiation Oncology, Ohio State University, Columbus, OH, USA
| | - Greg Krempl
- Department of Otolaryngology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - John A Ridge
- Department of Otolaryngology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - James A Bonner
- Department of Radiation Oncology, University of Alabama at Birmingham Medical Center, Birmingham, AL, USA
| | - Edith Filion
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Neal E Dunlap
- Department of Radiation Oncology, The James Graham Brown Cancer Center at University of Louisville, Louisville, KY, USA
| | - William A Stokes
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Michelle Mierzwa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew J Schipper
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA; Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
38
|
Jin Y, Ren H, Yue Q, Wu W, Liu C, Guo Y, Zhao P. Surrogacy of one-year survival for overall survival in advanced hepatocellular carcinoma. BMC Cancer 2024; 24:258. [PMID: 38395854 PMCID: PMC10893652 DOI: 10.1186/s12885-024-12000-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The increasing number of sequential treatments complicates the evaluation of overall survival (OS) in clinical trials for hepatocellular carcinoma (HCC), therefore, reliable surrogate endpoints (SEs) are required. This study aimed to evaluate the surrogacy of progression-free survival (PFS) and one-year (1-yr) milestone survival for OS in HCC trials. METHODS We systematically searched databases for randomized clinical trials that evaluated systemic treatments for advanced HCC. Individual patient data were reconstructed to calculate the 1-yr survival rate. We adopted a two-stage meta-analytic validation model to evaluate the correlation between SEs and OS, and the correlation between treatment effects on SEs and OS. The hazard ratio (HR) was calculated to assess the treatment effects on PFS and OS, and the 1-yr survival ratio was calculated to evaluate the treatment effects on the 1-yr milestone survival. RESULTS Thirty-two HCC trials involving 13,808 patients were included. A weak correlation was detected between the median PFS and median OS (R2 = 0.32), whereas the correlation improved between PFS HR and OS HR (R2 = 0.58). We identified strong correlations between the 1-yr survival rate and median OS and between the 1-yr survival ratio and OS HR (R2 = 0.74 and 0.65, respectively). In subgroup analyses, PFS HR strongly correlated with OS HR in trials relevant to immune checkpoint inhibitors (ICIs). Although the correlation remained weak between PFS and OS even in trials with PFS HR ≤ 0.6, the 1-yr survival rate and 1-yr survival ratio were strong surrogates for median OS and OS HR, respectively (R2 = 0.77 and 0.75). CONCLUSIONS One-year milestone survival outperformed PFS as a SE for OS in HCC, indicating the application of 1-yr survival as a secondary endpoint. In particular, PFS HR was a potential SE for OS HR in the ICI trials.
Collapse
Affiliation(s)
- Yuzhi Jin
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University & National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Hui Ren
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University & National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Qianhua Yue
- Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University & National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Chuan Liu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University & National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Yixuan Guo
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University & National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University & National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
- The First Affiliated Hospital, Zhejiang University School of Medicine & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 79 Qingchun Road, Hangzhou, China.
| |
Collapse
|
39
|
Wang Z, Zheng C, Han X, Chen W, Lu L. An Innovative and Efficient Diagnostic Prediction Flow for Head and Neck Cancer: A Deep Learning Approach for Multi-Modal Survival Analysis Prediction Based on Text and Multi-Center PET/CT Images. Diagnostics (Basel) 2024; 14:448. [PMID: 38396486 PMCID: PMC10888043 DOI: 10.3390/diagnostics14040448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Objective: To comprehensively capture intra-tumor heterogeneity in head and neck cancer (HNC) and maximize the use of valid information collected in the clinical field, we propose a novel multi-modal image-text fusion strategy aimed at improving prognosis. Method: We have developed a tailored diagnostic algorithm for HNC, leveraging a deep learning-based model that integrates both image and clinical text information. For the image fusion part, we used the cross-attention mechanism to fuse the image information between PET and CT, and for the fusion of text and image, we used the Q-former architecture to fuse the text and image information. We also improved the traditional prognostic model by introducing time as a variable in the construction of the model, and finally obtained the corresponding prognostic results. Result: We assessed the efficacy of our methodology through the compilation of a multicenter dataset, achieving commendable outcomes in multicenter validations. Notably, our results for metastasis-free survival (MFS), recurrence-free survival (RFS), overall survival (OS), and progression-free survival (PFS) were as follows: 0.796, 0.626, 0.641, and 0.691. Our results demonstrate a notable superiority over the utilization of CT and PET independently, and exceed the result derived without the clinical textual information. Conclusions: Our model not only validates the effectiveness of multi-modal fusion in aiding diagnosis, but also provides insights for optimizing survival analysis. The study underscores the potential of our approach in enhancing prognosis and contributing to the advancement of personalized medicine in HNC.
Collapse
Affiliation(s)
- Zhaonian Wang
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
| | - Chundan Zheng
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
| | - Xu Han
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
| | - Wufan Chen
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
| | - Lijun Lu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
| |
Collapse
|
40
|
Solanki AA, Yoo RK, Adams W, Davicioni E, Mysz ML, Shea S, Gupta GN, Showalter T, Garant A, Hentz C, Farooq A, Baldea K, Small W, Harkenrider MM. F-SHARP: a Phase I/II trial of focal salvage high-dose-rate brachytherapy for Radiorecurrent prostate cancer. BJU Int 2024; 133:188-196. [PMID: 37562825 DOI: 10.1111/bju.16150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
BACKGROUND Intraprostatic local radiorecurrence (LRR) after definitive radiation is being increasingly identified due to the implementation of molecular positron emission tomography (PET)/computed tomography (CT) imaging for the evaluation of biochemical recurrence. Salvage high-dose rate (HDR) brachytherapy offers a promising local therapy option, with encouraging toxicity and efficacy based on early series. Furthermore, the incorporation of advanced imaging allows for focal HDR to further reduce toxicity to maximise the therapeutic ratio. The objectives of the 'focal salvage HDR brachytherapy for locally recurrent prostate cancer in patients treated with prior radiotherapy' (F-SHARP) trial are to determine the acute and late toxicity and efficacy outcomes of focal salvage HDR brachytherapy for LRR prostate cancer. STUDY DESIGN The F-SHARP is a multi-institutional two-stage Phase I/II clinical trial of salvage focal HDR brachytherapy for LRR prostate cancer enrolling patients at three centres. ENDPOINTS The primary endpoint is the acute radiation-related Grade ≥3 Common Terminology Criteria for Adverse Events (CTCAE, version 4.03) genitourinary (GU) and gastrointestinal (GI) toxicity rate, defined as within 3 months of brachytherapy. Secondary endpoints include acute and late CTCAE toxicity, biochemical failure, patterns of clinical progression, disease-specific and overall survival, and health-related quality of life, as measured by the International Prostate Symptom Score and 26-item Expanded Prostate Cancer Index Composite instruments. PATIENTS AND METHODS Key eligibility criteria include: biopsy confirmed LRR prostate adenocarcinoma after prior definitive radiation therapy using any radiotherapeutic modality, no evidence of regional or distant metastasis, and cT1-3a Nx or N0 prostate cancer at initial treatment. All patients will have multiparametric magnetic resonance imaging and molecular PET/CT imaging if possible. In Stage 1, seven patients will be accrued. If there are two or more GI or GU Grade ≥3 toxicities, the study will be stopped. Otherwise, 17 additional patients will be accrued (total of 24 patients). For Stage 2, the cohort will expand to 62 subjects to study the efficacy outcomes, long-term toxicity profile, quality of life, and compare single- vs multi-fraction HDR. Transcriptomic analysis of recurrence biopsies will be performed to identify potential prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Abhishek A Solanki
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
- Department of Radiation Oncology, Loyola University Medical Center, Maywood, IL, USA
| | - Ryan K Yoo
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
- Department of Radiation Oncology, Loyola University Medical Center, Maywood, IL, USA
| | - William Adams
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL, USA
| | | | - Michael L Mysz
- Department of Radiation Oncology, Loyola University Medical Center, Maywood, IL, USA
| | - Steven Shea
- Department of Radiology, Stritch School of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL, USA
| | - Gopal N Gupta
- Department of Urology, Stritch School of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL, USA
| | - Timothy Showalter
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA
| | - Aurelie Garant
- Department of Radiation Oncology, University of Texas Southwestern, Dallas, TX, USA
| | | | - Ahmer Farooq
- Department of Urology, Stritch School of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL, USA
| | - Kristin Baldea
- Department of Urology, Stritch School of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL, USA
| | - William Small
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
- Department of Radiation Oncology, Loyola University Medical Center, Maywood, IL, USA
| | - Matthew M Harkenrider
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
- Department of Radiation Oncology, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
41
|
Terlizzi M, Bossi A. ProtecT trial: What's new after 15 years of follow-up: Re: Hamdy FC and colleagues; N Engl J Med. 2023 Apr 27. Urologia 2024; 91:3-4. [PMID: 38477298 DOI: 10.1177/03915603231225604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Recent results of ProtecT trial published after 15 years of follow-up demonstrate the absence of difference in prostate cancer-specific survival between active monitoring, radiotherapy, or prostatectomy for PSA-detected, localized prostate cancer patients. These results definitively confirm the essential role of active surveillance as the gold standard for men with low-risk and highly selected intermediate-risk prostate cancer. It underlines the importance of shared-decision making process with patients.
Collapse
Affiliation(s)
- Mario Terlizzi
- Radiation Oncology Department, Institut Gustave Roussy, Villejuif, France
| | - Alberto Bossi
- Radiation Oncology Department, Institut Gustave Roussy, Villejuif, France
- Amethyst Radiotherapy Group, La Garenne Colombes, France
| |
Collapse
|
42
|
Childs DS, Orme JJ, Ravi P. Delaying Prostate-Specific Antigen Progression in Biochemically Recurrent Prostate Cancer: Is It Clinically Meaningful? J Clin Oncol 2024:JCO2302410. [PMID: 38261976 DOI: 10.1200/jco.23.02410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 01/25/2024] Open
Affiliation(s)
| | - Jacob J Orme
- Department of Oncology, Mayo Clinic, Rochester, MN
| | - Praful Ravi
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
43
|
McKone EL, Sutton EA, Johnson GB, Phillips RM. Application of Advanced Imaging to Prostate Cancer Diagnosis and Management: A Narrative Review of Current Practice and Unanswered Questions. J Clin Med 2024; 13:446. [PMID: 38256579 PMCID: PMC10816977 DOI: 10.3390/jcm13020446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Major advances in prostate cancer diagnosis, staging, and management have occurred over the past decade, largely due to our improved understanding of the technical aspects and clinical applications of advanced imaging, specifically magnetic resonance imaging (MRI) and prostate-cancer-specific positron emission tomography (PET). Herein, we review the established utility of these important and exciting technologies, as well as areas of controversy and uncertainty that remain important areas for future study. There is strong evidence supporting the utility of MRI in guiding initial biopsy and assessing local disease. There is debate, however, regarding how to best use the imaging modality in risk stratification, treatment planning, and assessment of biochemical failure. Prostate-cancer-specific PET is a relatively new technology that provides great value to the evaluation of newly diagnosed, treated, and recurrent prostate cancer. However, its ideal use in treatment decision making, staging, recurrence detection, and surveillance necessitates further research. Continued study of both imaging modalities will allow for an improved understanding of their best utilization in improving cancer care.
Collapse
Affiliation(s)
| | - Elsa A. Sutton
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Geoffrey B. Johnson
- Department of Radiology, Nuclear Medicine Division, Mayo Clinic, Rochester, MN 55905, USA
| | - Ryan M. Phillips
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
44
|
Sumiyoshi T, Wang X, Warner EW, Sboner A, Annala M, Sigouros M, Beja K, Mizuno K, Ku S, Fazli L, Eastham J, Taplin ME, Simko J, Halabi S, Morris MJ, Gleave ME, Wyatt AW, Beltran H. Molecular features of prostate cancer after neoadjuvant therapy in the phase 3 CALGB 90203 trial. J Natl Cancer Inst 2024; 116:115-126. [PMID: 37676819 PMCID: PMC10777679 DOI: 10.1093/jnci/djad184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/31/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND The phase 3 CALGB 90203 (Alliance) trial evaluated neoadjuvant chemohormonal therapy for high-risk localized prostate cancer before radical prostatectomy. We dissected the molecular features of post-treated tumors with long-term clinical outcomes to explore mechanisms of response and resistance to chemohormonal therapy. METHODS We evaluated 471 radical prostatectomy tumors, including 294 samples from 166 patients treated with 6 cycles of docetaxel plus androgen deprivation therapy before radical prostatectomy and 177 samples from 97 patients in the control arm (radical prostatectomy alone). Targeted DNA sequencing and RNA expression of tumor foci and adjacent noncancer regions were analyzed in conjunction with pathologic changes and clinical outcomes. RESULTS Tumor fraction estimated from DNA sequencing was significantly lower in post-treated tumor tissues after chemohormonal therapy compared with controls. Higher tumor fraction after chemohormonal therapy was associated with aggressive pathologic features and poor outcomes, including prostate-specific antigen-progression-free survival. SPOP alterations were infrequently detected after chemohormonal therapy, while TP53 alterations were enriched and associated with shorter overall survival. Residual tumor fraction after chemohormonal therapy was linked to higher expression of androgen receptor-regulated genes, cell cycle genes, and neuroendocrine genes, suggesting persistent populations of active prostate cancer cells. Supervised clustering of post-treated high-tumor-fraction tissues identified a group of patients with elevated cell cycle-related gene expression and poor clinical outcomes. CONCLUSIONS Distinct recurrent prostate cancer genomic and transcriptomic features are observed after exposure to docetaxel and androgen deprivation therapy. Tumor fraction assessed by DNA sequencing quantifies pathologic response and could be a useful trial endpoint or prognostic biomarker. TP53 alterations and high cell cycle transcriptomic activity are linked to aggressive residual disease, despite potent chemohormonal therapy.
Collapse
Affiliation(s)
- Takayuki Sumiyoshi
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xiaofei Wang
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Evan W Warner
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Matti Annala
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kevin Beja
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Kei Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shengyu Ku
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ladan Fazli
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - James Eastham
- Urology Service at the Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jeffrey Simko
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke Cancer Institute, Durham, NC, USA
| | - Michael J Morris
- Department of Genitourinary Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin E Gleave
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
45
|
Valentin B, Arsov C, Ullrich T, Al-Monajjed R, Boschheidgen M, Hadaschik BA, Giganti F, Giessing M, Lopez-Cotarelo C, Esposito I, Antoch G, Albers P, Radtke JP, Schimmöller L. Magnetic Resonance Imaging-guided Active Surveillance Without Annual Rebiopsy in Patients with Grade Group 1 or 2 Prostate Cancer: The Prospective PROMM-AS Study. EUR UROL SUPPL 2024; 59:30-38. [PMID: 38298772 PMCID: PMC10829616 DOI: 10.1016/j.euros.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 02/02/2024] Open
Abstract
Background Multiparametric magnetic resonance imaging (mpMRI) may allow patients with prostate cancer (PC) on active surveillance (AS) to avoid repeat prostate biopsies during monitoring. Objective To assess the ability of mpMRI to reduce guideline-mandated biopsy and to predict grade group upgrading in patients with International Society of Urological Pathology grade group (GG) 1 or GG 2 PC using Prostate Cancer Radiological Estimation of Change in Sequential Evaluation (PRECISE) scores. The hypothesis was that the AS disqualification rate (ASDQ) rate could be reduced to 15%. Design setting and participants PROMM-AS was a prospective study assessing 2-yr outcomes for an mpMRI-guided AS protocol. A 12 mo after AS inclusion on the basis of MRI/transrectal ultrasound fusion-guided biopsy (FBx), all patients underwent mpMRI. For patients with stable mpMRI (PRECISE 1-3), repeat biopsy was deferred and follow-up mpMRI was scheduled for 12 mo later. Patients with mpMRI progression (PRECISE 4-5) underwent FBx. At the end of the study, follow-up FBx was indicated for all patients. Outcome measurements and statistical analysis We calculated the sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) for upgrading to GG 2 in the GG 1 group, and to GG 3 in the GG 2 group on MRI. We performed regression analyses that included clinical variables. Results and limitations The study included 101 patients with PC (60 GG 1 and 41 GG 2). Histopathological progression occurred in 31 patients, 18 in the GG 1 group and 13 in the GG 2 group. Thus, the aim of reducing the ASDQ rate to 15% was not achieved. The sensitivity, specificity, PPV, and NPV for PRECISE scoring of MRI were 94%, 64%, 81%, and 88% in the GG 1 group, and 92%, 50%, 92%, and 50%, respectively, in the GG 2 group. On regression analysis, initial prostate-specific antigen (p < 0.001) and higher PRECISE score (4-5; p = 0.005) were significant predictors of histological progression of GG 1 PC. Higher PRECISE score (p = 0.009), initial Prostate Imaging-Reporting and Data System score (p = 0.009), previous negative biopsy (p = 0.02), and percentage Gleason pattern 4 (p = 0.04) were significant predictors of histological progression of GG 2 PC. Limitations include extensive MRI reading experience, the small sample size, and limited follow-up. Conclusions MRI-guided monitoring of patients on AS using PRECISE scores avoided unnecessary follow-up biopsies in 88% of patients with GG 1 PC and predicted upgrading during 2-yr follow-up in both GG 1 and GG 2 PC. Patient summary We investigated whether MRI (magnetic resonance imaging) scores can be used to guide whether patients with lower-risk prostate cancer who are on active surveillance (AS) need to undergo repeat biopsies. Follow-up biopsy was deferred for 1 year for patients with a stable score and performed for patients whose score progressed. After 24 months on AS, all men underwent MRI and biopsy. Among patients with grade group 1 cancer and a stable MRI score, 88% avoided biopsy. For patients with MRI score progression, AS termination was correctly recommended in 81% of grade group 1 and 92% of grade group 2 cases.
Collapse
Affiliation(s)
- Birte Valentin
- Department of Diagnostic and Interventional Radiology, University of Düsseldorf, Düsseldorf, Germany
| | - Christian Arsov
- Department of Urology, University of Düsseldorf, Düsseldorf, Germany
- Elisabeth-Krankenhaus Rheydt, Department of Urology and Paediatric Urology, Staedtische Kliniken Moenchengladbach GmbH, Moenchengladbach, Germany
| | - Tim Ullrich
- Department of Diagnostic and Interventional Radiology, University of Düsseldorf, Düsseldorf, Germany
| | | | - Matthias Boschheidgen
- Department of Diagnostic and Interventional Radiology, University of Düsseldorf, Düsseldorf, Germany
| | | | - Francesco Giganti
- Department of Radiology, University College London Hospitals NHS Foundation Trust, London, UK
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Markus Giessing
- Department of Urology, University of Düsseldorf, Düsseldorf, Germany
| | | | - Irene Esposito
- Institute of Pathology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, University of Düsseldorf, Düsseldorf, Germany
| | - Peter Albers
- Department of Urology, University of Düsseldorf, Düsseldorf, Germany
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Jan Philipp Radtke
- Department of Urology, University of Düsseldorf, Düsseldorf, Germany
- Department of Radiology, German Cancer Research Center, Heidelberg, Germany
| | - Lars Schimmöller
- Department of Diagnostic and Interventional Radiology, University of Düsseldorf, Düsseldorf, Germany
- Department of Diagnostic, Interventional Radiology and Nuclear Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| |
Collapse
|
46
|
Roberts EK, Elliott MR, Taylor JMG. Surrogacy validation for time-to-event outcomes with illness-death frailty models. Biom J 2024; 66:e2200324. [PMID: 37776057 PMCID: PMC10873101 DOI: 10.1002/bimj.202200324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/20/2023] [Accepted: 06/15/2023] [Indexed: 10/01/2023]
Abstract
A common practice in clinical trials is to evaluate a treatment effect on an intermediate outcome when the true outcome of interest would be difficult or costly to measure. We consider how to validate intermediate outcomes in a causally-valid way when the trial outcomes are time-to-event. Using counterfactual outcomes, those that would be observed if the counterfactual treatment had been given, the causal association paradigm assesses the relationship of the treatment effect on the surrogate outcome with the treatment effect on the true, primary outcome. In particular, we propose illness-death models to accommodate the censored and semicompeting risk structure of survival data. The proposed causal version of these models involves estimable and counterfactual frailty terms. Via these multistate models, we characterize what a valid surrogate would look like using a causal effect predictiveness plot. We evaluate the estimation properties of a Bayesian method using Markov chain Monte Carlo and assess the sensitivity of our model assumptions. Our motivating data source is a localized prostate cancer clinical trial where the two survival outcomes are time to distant metastasis and time to death.
Collapse
Affiliation(s)
| | - Michael R. Elliott
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
- Survey Methodology Program, Institute for Social Research Ann Arbor, MI
| | | |
Collapse
|
47
|
González-Padilla DA, Subiela JD. Are PARP Inhibitors Ready for Prime Time in Metastatic Prostate Cancer? Maybe Not. Eur Urol 2024; 85:1-2. [PMID: 37244815 DOI: 10.1016/j.eururo.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/10/2023] [Indexed: 05/29/2023]
Abstract
While PARP inhibitors such as rucaparib and olaparib have shown activity in metastatic castration-resistant prostate cancer, they have failed to show a clear improvement in hard outcomes such as overall survival or quality of life. Because of methodological limitations, we suggest caution before implementing these treatments in routine clinical practice; offering them to patients without a BRCA1/2 mutation is probably inappropriate.
Collapse
Affiliation(s)
| | - José Daniel Subiela
- Department of Urology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Madrid, Spain
| |
Collapse
|
48
|
Gilbert DC, Nankivell M, Rush H, Clarke NW, Mangar S, Al-Hasso A, Rosen S, Kockelbergh R, Sundaram SK, Dixit S, Laniado M, McPhail N, Shaheen A, Brown S, Gale J, Deighan J, Marshall J, Duong T, Macnair A, Griffiths A, Amos CL, Sydes MR, James ND, Parmar MKB, Langley RE. A Repurposing Programme Evaluating Transdermal Oestradiol Patches for the Treatment of Prostate Cancer Within the PATCH and STAMPEDE Trials: Current Results and Adapting Trial Design. Clin Oncol (R Coll Radiol) 2024; 36:e11-e19. [PMID: 37973477 DOI: 10.1016/j.clon.2023.10.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
AIMS Androgen deprivation therapy (ADT), usually achieved with luteinising hormone releasing hormone analogues (LHRHa), is central to prostate cancer management. LHRHa reduce both testosterone and oestrogen and are associated with significant long-term toxicity. Previous use of oral oestrogens as ADT was curtailed because of cardiovascular toxicity. Transdermal oestrogen (tE2) patches are a potential alternative ADT, supressing testosterone without the associated oestrogen-depletion toxicities (osteoporosis, hot flushes, metabolic abnormalities) and avoiding cardiovascular toxicity, and we here describe their evaluation in men with prostate cancer. MATERIALS AND METHODS The PATCH (NCT00303784) adaptive trials programme (incorporating recruitment through the STAMPEDE [NCT00268476] platform) is evaluating the safety and efficacy of tE2 patches as ADT for men with prostate cancer. An initial randomised (LHRHa versus tE2) phase II study (n = 251) with cardiovascular toxicity as the primary outcome measure has expanded into a phase III evaluation. Those with locally advanced (M0) or metastatic (M1) prostate cancer are eligible. To reflect changes in both management and prognosis, the PATCH programme is now evaluating these cohorts separately. RESULTS Recruitment is complete, with 1362 and 1128 in the M0 and M1 cohorts, respectively. Rates of androgen suppression with tE2 were equivalent to LHRHa, with improved metabolic parameters, quality of life and bone health indices (mean absolute change in lumbar spine bone mineral density of -3.0% for LHRHa and +7.9% for tE2 with an estimated difference between arms of 9.3% (95% confidence interval 5.3-13.4). Importantly, rates of cardiovascular events were not significantly different between the two arms and the time to first cardiovascular event did not differ between treatment groups (hazard ratio 1.11, 95% confidence interval 0.80-1.53; P = 0.54). Oncological outcomes are awaited. FUTURE Efficacy results for the M0 cohort (primary outcome measure metastases-free survival) are expected in the final quarter of 2023. For M1 patients (primary outcome measure - overall survival), analysis using restricted mean survival time is being explored. Allied translational work on longitudinal samples is underway.
Collapse
Affiliation(s)
- D C Gilbert
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK; University Hospitals Sussex NHS Foundation Trust, Royal Sussex County Hospital, Brighton, UK.
| | - M Nankivell
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - H Rush
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - N W Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - S Mangar
- Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - A Al-Hasso
- Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - S Rosen
- National Heart and Lung Institute, Imperial College, London, UK
| | - R Kockelbergh
- Department of Urology, University Hospitals of Leicester, Leicester, UK
| | - S K Sundaram
- Mid-Yorkshire Teaching NHS Trust, Pinderfields Hospital, Wakefield, UK
| | - S Dixit
- Scunthorpe General Hospital, Scunthorpe, UK
| | | | | | | | - S Brown
- Airedale General Hospital, Keighley, UK
| | - J Gale
- Queen Alexandra Hospital, Portsmouth, UK
| | - J Deighan
- Patient Representative, MRC Clinical Trials Unit at UCL, London, UK
| | - J Marshall
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - T Duong
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - A Macnair
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK; Guys and St Thomas' NHS Foundation Trust, London, UK
| | - A Griffiths
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - C L Amos
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - M R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - N D James
- Institute of Cancer Research, Sutton, UK
| | - M K B Parmar
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| | - R E Langley
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, London, UK
| |
Collapse
|
49
|
Shahnam A, Nindra U, Desai J, Hui R, Buyse M, Hopkins AM, Sorich MJ. Time to deterioration of patient-reported outcomes as a surrogate of overall survival: a meta-analysis. J Natl Cancer Inst 2023; 115:1475-1482. [PMID: 37540222 DOI: 10.1093/jnci/djad152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/06/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Overall survival is the optimal marker of treatment efficacy in randomized clinical trials (RCTs) but can take considerable time to mature. Progression-free survival (PFS) has served as an early surrogate of overall survival but is imperfect. Time to deterioration in quality of life (QOL) measures could be a surrogate for overall survival. METHODS Phase 3 RCTs in solid malignancies that reported overall survival, PFS, and time to deterioration in QOL or physical function published between January 1, 2010, and June 30, 2022, were evaluated. Weighted regression analysis was used to assess the relationship between PFS, time to deterioration in QOL, and time to deterioration in physical function with overall survival. The coefficient of determination (R2) was used to quantify surrogacy. RESULTS In total, 138 phase 3 RCTs were included. Of these, 47 trials evaluated immune checkpoint inhibitors and 91 investigated non-immune checkpoint inhibitor agents. Time to deterioration in QOL (137 RCTs) and time to deterioration in physical function (75 RCTs) performed similarly to PFS as surrogates for overall survival (R2 = 0.18 vs R2 = 0.19 and R2 = 0.10 vs R2 = 0.09, respectively). For immune checkpoint inhibitor studies, time to deterioration in physical function had a higher association with overall survival than with PFS (R2 = 0.38 vs R2 = 0.19), and PFS and time to deterioration in physical function did not correlate with each other (R2 = 0). When time to deterioration in physical function and PFS are used together, the coefficient of determination increased (R2 = 0.57). CONCLUSIONS Time to deterioration in physical function appears to be an overall survival surrogate measure of particular importance for immune checkpoint inhibitor treatment efficacy. The combination of time to deterioration in physical function with PFS may enable better prediction of overall survival treatment benefit in RCTs of immune checkpoint inhibitors than either PFS or time to deterioration in physical function alone.
Collapse
Affiliation(s)
- Adel Shahnam
- Department of Medical Oncology, Peter McCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Udit Nindra
- Department of Medical Oncology, Liverpool Hospital, Sydney, New South Wales, Australia
| | - Jayesh Desai
- Department of Medical Oncology, Peter McCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Rina Hui
- Centre of Cancer Medicine, University of Hong Kong, Hong Kong
| | - Marc Buyse
- International Drug Development Institute, Brussels, Belgium
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
50
|
Milenkovic U, Kuijk J, Roussel E, Devos G, Van den Broeck T, Van Eecke H, Vanderstichele A, Duvillier T, Verhamme L, Van Haute W, Goeman L, Berghen C, Joniau S, De Meerleer G. Predictors of Recurrence After Metastasis-directed Therapy in Oligorecurrent Prostate Cancer Following Radical Prostatectomy. Eur Urol Oncol 2023; 6:582-589. [PMID: 36878753 DOI: 10.1016/j.euo.2023.02.010] [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: 03/23/2022] [Revised: 01/12/2023] [Accepted: 02/16/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Metastasis-directed therapy (MDT) is performed to delay systemic treatments for oligorecurrent disease after primary prostate cancer (PCa) treatment. OBJECTIVE The aim of this study was to identify the predictors of therapeutic response of MDT for oligorecurrent PCa. DESIGN, SETTING, AND PARTICIPANTS bicentric, retrospective study, including consecutive patients who underwent MDT for oligorecurrent PCa after radical prostatectomy (RP; 2006-2020) was conducted. MDT encompassed stereotactic body radiation therapy (SBRT), salvage lymph node dissection (sLND), whole-pelvis/retroperitoneal radiation therapy (WP[R]RT), or metastasectomy. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS ndpoints were 5-yr radiographic progression-free survival (rPFS), metastasis-free survival (MFS), palliative androgen deprivation treatment (pADT)-free survival, and overall survival (OS) together with prognostic factors for MFS following primary MDT. Survival outcomes were studied by Kaplan-Meier survival and univariable Cox regression (UVA). RESULTS AND LIMITATIONS A total of 211 MDT patients were included; 122 (58%) developed a secondary recurrence. Salvage lymph node dissection was performed in 119 (56%), SBRT in 48 (23%), and WP(R)RT in 31 (15%) of the cases. Two patients received sLND + SBRT and one received sLND + WPRT. Eleven (5%) patients received metastasectomies. The median follow-up since RP was 100 mo, while follow-up after MDT was 42 mo. The 5-yr rPFS, MFS, androgen deprivation treatment(-free survival, castration-resistant prostate cancer-free survival, CSS, and OS after MDT were 23%, 68%, 58%, 82%, 93%, and 87% respectively. There was a statistically significant difference between cN1 (n = 114) and cM+ (n = 97) for 5-yr MFS (83% vs 51%, p < 0.001), pADT-free survival (70% vs 49%, p = 0.014), and CSS (100% vs 86%, p = 0.019). UVA was performed to assess the risk factors (RFs) for MFS in cN1 and cM+. Alpha was set at 10%. RFs for MFS in cN1 were lower initial prostate-specific antigen (PSA) at the time of RP (hazard ratio [95% confidence interval] 0.15 [0.02-1.02], p = 0.053], pN stage at RP (2.91 [0.83-10.24], p = 0.096), nonpersisting PSA after RP (0.47 [0.19-1.12], p = 0.089), higher PSA at primary MDT (2.38 [1.07-5.24], p = 0.032), and number of positive nodes on imaging (1.65 [1.14-2.40], p < 0.01). RFs for MFS in cM+ were higher pathological Gleason score (1.86 [0.93-3.73], p = 0.078), number of lesions on imaging (0.77 [0.57-1.04], p = 0.083), and cM1b/cM1c (non-nodal metastatic recurrence; 2.62 [1.58-4.34], p < 0.001). CONCLUSIONS Following MDT, 23% of patients were free of a second recurrence at 5-yr follow-up. Moreover, cM+ patients had significantly worse outcomes in terms of MFS, pADT-free survival, and CSS. The RFs for a metastatic recurrence can be used for counseling patients, to inform prognosis, and potentially select candidates for MDT. PATIENT SUMMARY In this paper, we looked at the outcomes of using localized, patient-tailored treatment for imaging-detected recurrent prostate cancer in lymph nodes, bone, or viscera (maximum five recurrences on imaging). Our results showed that targeted treatment of the metastatic lesions could delay the premature use of hormone therapy.
Collapse
Affiliation(s)
- Uros Milenkovic
- Department of Urology, AZ Delta, Roeselare, Belgium; Department of Urology, University Hospitals Leuven, Leuven, Belgium.
| | - Joke Kuijk
- Department of Urology, AZ Delta, Roeselare, Belgium
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Gaetan Devos
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | | | - Henri Van Eecke
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | | | | | | | | | | | - Charlien Berghen
- Department of Radiotherapy, University Hospitals Leuven, Leuven, Belgium
| | - Steven Joniau
- Department of Urology, AZ Delta, Roeselare, Belgium; Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Gert De Meerleer
- Department of Radiotherapy, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|