1
|
Roidos C, Anastasiadis A, Tsiakaras S, Loutradis C, Baniotis P, Memmos D, Dimitriadis G, Papaioannou M. Integration of Genomic Tests in Prostate Cancer Care: Implications for Clinical Practice and Patient Outcomes. Curr Issues Mol Biol 2024; 46:14408-14421. [PMID: 39727992 DOI: 10.3390/cimb46120864] [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: 10/30/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Prostate cancer (PCa) is a common malignancy in men and is among the leading causes of cancer-related death worldwide. Genomic tests assess disease aggressiveness and guide treatment, particularly in low- and intermediate-risk PCa. We reviewed the literature on the use of four genomic tests (Prolaris®, Promark®, Oncotype DX®, and Decipher®) in assessing the prognosis of PCa and their use in treatment decision-making. Most of the studies showed that Prolaris® has a strong correlation with biochemical recurrence, metastasis risk, PCa-specific mortality (PCSM), and pathological features. Similarly, three studies on Promark® indicated a connection between results and pathological features in the subsequent prostatectomy, time to metastasis, and biochemical recurrence. Fourteen studies on Oncotype DX® showed a clear correlation between high scores, death, and PCSM. One study found that routine biopsy pathology reports, combined with serum PSA levels, provide a risk assessment comparable to Oncotype DX® testing. Results from 22 studies on Decipher® were controversial. The test was associated with conservative management, suggesting that patients with a high GC score are more likely to need radiation after surgery. Comparative studies indicated that Oncotype DX® is preferable for assessing PCSM, Decipher® for predicting metastasis, and Prolaris® for predicting recurrence. With the incidence rate of PCa dramatically increasing, genomic tests appear to be useful adjunctive precision medicine tools with significant potential in improving prognostic discrimination, facilitating better risk stratification, and guiding personalized treatment, especially in the intermediate-risk patient group. Large-scale, prospective, multi-sectional studies are required to validate the utility of these tests prior to their integration into clinical practice.
Collapse
Affiliation(s)
- Christos Roidos
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Anastasios Anastasiadis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Stavros Tsiakaras
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Charalampos Loutradis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Panagiotis Baniotis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Dimitrios Memmos
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Georgios Dimitriadis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Maria Papaioannou
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
- Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| |
Collapse
|
2
|
Nourmohammadi Abadchi S, Salles DC, Flannery C, Sama V, Baehner FL, Zambon JP, Mendes AA, DePaula Oliveira L, Han M, Jing Y, Partin AW, Trock BJ, Lotan TL. Association of Genomic Prostate Score at positive margin with recurrence after radical prostatectomy. BJU Int 2024; 134:939-944. [PMID: 38953550 DOI: 10.1111/bju.16445] [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] [Indexed: 07/04/2024]
Abstract
OBJECTIVES To evaluate the utility of the 17-gene Genomic Prostate Score® (GPS; MDxHealth, Irvine, CA, USA) performed on prostate cancer at the positive margin of the radical prostatectomy (RP) for its association with risk of subsequent biochemical recurrence (BCR). PATIENTS AND METHODS We designed a case-cohort for the outcome of BCR, selecting 223 from a cohort of 813 RP patients treated at Johns Hopkins from 2008 to 2017 with positive margins and available clinical data; of these, 213 had available tissue and clinical data. RNA was isolated from formalin-fixed paraffin-embedded tumour tissue adjacent to the positive surgical margin and the GPS was evaluable in 203 of these patients with a score ranging from 0 to 100, with higher scores indicating higher risk. All patients underwent RP with or without adjuvant radiation therapy (ART). The statistical analysis employed Cox proportional hazards regression models for outcome of BCR weighted for case-cohort design. RESULTS In univariable analysis, every 20-unit increase in the GPS was associated with a nearly threefold increase in risk of BCR (hazard ratio [HR] per 20 units 2.82, P < 0.001). In a multivariable Cox model adjusted for age, race, Cancer of the Prostate Risk Assessment Postsurgical score, Grade Group at the positive margin, and ART, the GPS was significantly associated with BCR (HR 1.56 per 20 units; 95% confidence interval 1.11-2.19; P = 0.011). The study is limited by its retrospective and single institution design. CONCLUSIONS The GPS at the positive surgical margin could help stratify prognosis and inform clinical decision-making regarding adjuvant therapy after RP.
Collapse
Affiliation(s)
| | - Daniela C Salles
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | - Adrianna A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lia DePaula Oliveira
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Misop Han
- MDX Health Corporation, Irvine, CA, USA
| | - Yuezhou Jing
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan W Partin
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruce J Trock
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Lee E, Oliveira LD, Dairo O, Nourmohammadi Abadchi S, Cha E, Mendes AA, Wang JH, Song DY, Lotan TL. PTEN Loss Is Associated with Adverse Outcomes in the Setting of Salvage Radiation Therapy. Eur Urol Oncol 2024; 7:1513-1519. [PMID: 38964997 DOI: 10.1016/j.euo.2024.06.008] [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: 02/21/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Salvage radiation therapy (SRT) is a mainstay of treatment for biochemical relapse following radical prostatectomy; however, few studies have examined genomic biomarkers in this context. OBJECTIVE We characterized the prognostic impact of previously identified deleterious molecular phenotypes-loss of PTEN, ERG expression, and TP53 mutation-for patients undergoing SRT. DESIGN, SETTING, AND PARTICIPANTS We leveraged an institutional database of 320 SRT patients with available tissue and follow-up. Tissue microarrays were used for genetically validated immunohistochemistry assays. INTERVENTION All men underwent SRT with or without androgen deprivation therapy OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Univariable and multivariable Cox-proportional hazard models assessed the association of molecular phenotypes with biochemical recurrence-free (bRFS) and metastasis-free (MFS) survival after SRT. RESULTS AND LIMITATIONS Loss of PTEN (n = 123, 43%) and ERG expression (n = 118, 39%) were common in this cohort, while p53 overexpression (signifying TP53 missense mutation) was infrequent (n = 21, 7%). In univariable analyses, any loss of PTEN portended worse bRFS (hazard ratio [HR] 1.86; 95% confidence interval 1.36-2.57) and MFS (HR 1.89; 1.21-2.94), with homogeneous PTEN loss being associated with the highest risk of MFS (HR 2.47; 1.54-3.95). Similarly, p53 overexpression predicted worse bRFS (HR 1.95; 1.14-3.32) and MFS (HR 2.79; 1.50-5.19). ERG expression was associated with worse MFS only (HR 1.6; 1.03-2.48). On the multivariable analysis adjusting for known prognostic features, homogeneous PTEN loss remained predictive of adverse bRFS (HR 1.82; 1.12-2.96) and MFS (HR 2.08; 1.06-4.86). The study is limited by its retrospective and single-institution design. CONCLUSIONS PTEN loss by immunohistochemistry is an independent adverse prognostic factor for bRFS and MFS in prostate cancer patients treated with SRT. Future trials will determine the optimal approach to treating SRT patients with adverse molecular prognostic features. PATIENT SUMMARY Loss of the PTEN tumor suppressor protein is associated with worse outcomes after salvage radiotherapy, independent of other clinical or pathologic patient characteristics.
Collapse
Affiliation(s)
- Emerson Lee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lia DePaula Oliveira
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Oluwademilade Dairo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Eumee Cha
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrianna A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jarey H Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel Y Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Hamid AA, Sweeney CJ, Hovens C, Corcoran N, Azad AA. Precision medicine for prostate cancer: An international perspective. Urol Oncol 2024; 42:392-401. [PMID: 38614920 DOI: 10.1016/j.urolonc.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 04/15/2024]
Abstract
Greater personalization of cancer medicine continues to shape therapy development and patient selection accordingly. The treatment of prostate cancer has evolved considerably since the discovery of androgen deprivation therapy. The comprehensive profiling of the prostate cancer genome has mapped the targetable molecular landscape of the disease and identified opportunities for the implementation of novel and combination therapies. In this review, we provide an overview of the molecular biology of prostate cancer and tools developed to aid prognostication and prediction of therapy benefit. Modern treatment of advanced prostate cancer is reviewed as a paradigm of increasing precision-informed approach to patient care, and must be considered on a global scale with respect to the state of science and care delivery.
Collapse
Affiliation(s)
- Anis A Hamid
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Surgery, University of Melbourne, Melbourne, Australia.
| | | | | | - Niall Corcoran
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Arun A Azad
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
5
|
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
|
6
|
Morgan TM, Daignault-Newton S, Spratt DE, Dunn RL, Singhal U, Okoth LA, Feng FY, Johnson AM, Lane BR, Linsell S, Ghani KR, Montie JE, Mehra R, Hollenbeck BK, Maatman T, Wojno K, Burks FN, Bekong D, Curry J, Rodriguez P, Kleer E, Sarle R, Miller DC, Cher ML. Impact of Gene Expression Classifier Testing on Adjuvant Treatment Following Radical Prostatectomy: The G-MINOR Prospective Randomized Cluster-crossover Trial. Eur Urol 2024:S0302-2838(24)02604-6. [PMID: 39379238 DOI: 10.1016/j.eururo.2024.09.011] [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: 03/01/2024] [Revised: 07/30/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND AND OBJECTIVE Decipher is a tissue-based genomic classifier (GC) developed and validated in the post-radical prostatectomy (RP) setting as a predictor of metastasis. We conducted a prospective randomized controlled cluster-crossover trial assessing the use of Decipher to determine its impact on adjuvant treatment after RP. METHODS Eligible patients had undergone RP within 9 mo of enrollment, had pT3-4 disease and/or positive surgical margins, and prostate-specific antigen <0.1 ng/ml. Centers were randomized to a sequence of 3-mo periods of either GC-informed care or usual care (UC). Cancer of the Prostate Risk Assessment Postsurgical (CAPRA-S) recurrence risk scores were provided to treating physicians and patients in all periods. KEY FINDINGS AND LIMITATIONS Impact of GC test results on adjuvant treatment were compared with UC alone. Longitudinal patient-reported urinary and sexual function was assessed. A total of 175 patients were enrolled in 27 periods with GC and 163 in 28 periods with UC. At 18 mo after RP, an average patient in the GC arm received adjuvant treatment 9.7% of the time compared with 8.7% for an average individual in the UC arm (0.99% mean difference, 95% confidence interval [CI] -7.6%, 9.6%, p = 0.8). While controlling for CAPRA-S score, higher GC scores tended to result in an increased likelihood of adjuvant treatment that was not statistically significant (odds ratio [OR] = 1.35 per 0.1 increase in GC score, 95% CI 0.98-1.85, p = 0.066). Using the GC risk groups, reflecting clinical use, a high GC risk was associated with significantly higher odds of receiving adjuvant treatment (OR = 6.9, 95% CI 1.8, 26, p = 0.005) compared with a low GC score, adjusted for CAPRA-S score. There were no differences in patient-reported urinary and sexual function between the study arms. As oncologic outcomes are immature, the present data cannot address whether GC testing provides any cancer control benefit. CONCLUSIONS AND CLINICAL IMPLICATIONS GC testing impacts adjuvant therapy administration when viewed through the risk categories presented in the patient report; however, these data do not provide specific support for GC testing in the adjuvant treatment setting.
Collapse
Affiliation(s)
| | | | - Daniel E Spratt
- University of Michigan, Ann Arbor, MI, USA; UH Seidman Cancer Center/Case Western Reserve University, Cleveland, OH, USA
| | - Rod L Dunn
- University of Michigan, Ann Arbor, MI, USA
| | | | | | - Felix Y Feng
- University of California San Francisco, San Francisco, CA, USA
| | | | - Brian R Lane
- Spectrum Health Medical Group, Grand Rapids, MI, USA
| | | | | | | | | | | | | | - Kirk Wojno
- Comprehensive Urology, Royal Oak, MI, USA
| | | | | | - Jon Curry
- Urologic Consultants P.C, Grand Rapids, MI USA
| | - Paul Rodriguez
- Urology Associates of Grand Rapids PC, Grand Rapids, MI, USA
| | | | | | | | | |
Collapse
|
7
|
Harsini S, Martineau P, Plaha S, Saprunoff H, Chen C, Bishop J, Tyldesley S, Wilson D, Bénard F. Prognostic significance of a negative PSMA PET/CT in biochemical recurrence of prostate cancer. Cancer Imaging 2024; 24:117. [PMID: 39210431 PMCID: PMC11363643 DOI: 10.1186/s40644-024-00752-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) is becoming standard of care for men with biochemical recurrence (BCR) of prostate cancer. The implications of a negative PSMA PET/CT scan in this population remain unclear. This study aims to assess the outcome of patients with BCR post radical prostatectomy (RP) who have negative [18F]DCFPyL PET/CT scan at relapse. METHODS This is a post-hoc subgroup analysis of a prospective non randomized clinical trial. One hundred and one patients (median age, 75 years) with BCR after RP, who tested negative on [18F]DCFPyL PET/CT and subsequently either underwent salvage radiotherapy (sRT) with or without androgen deprivation therapy (ADT) or were followed without active treatment, were included. Freedom from progression (FFP) after negative PSMA PET/CT was determined based on follow-up imaging selected as per clinical practice. Uni- and multivariate Cox regression analyses were performed to examine the association of patients' characteristics, tumor-specific variables, and treatment with clinical progression at the last follow-up. FFP at 1-, 2-, and 3-year were reported using Kaplan Meier analysis. RESULTS The median PSA level at PET/CT was 0.56 ng/mL (range, 0.4-11.3). Sixty five (64%) patients were followed without receiving further treatment, and 36 (36%) received sRT (18% to the prostate bed only and 18% to the prostate bed and pelvic lymph nodes) within 3 months of the PSMA PET. Seventeen of the sRT patients (17 of 36, 47%) received concomitant androgen deprivation therapy (ADT). Median follow-up was 39 months. Subsequent clinical progression was detected in 21 patients (21%), with 52% in pelvic lymph nodes, 52% in the prostatic fossa, 19% in distant lymph nodes, 14% in lungs, and 10% in bones. The FFP was 95% (95% CI: 91%-99%) at 12 months, 87% (95% CI: 81%-94%) at 24 months, and 79% (95% CI: 71%-88%) at 36 months. Multivariate Cox regression analysis revealed that an initial International Society of Urological Pathology (ISUP) grade 5 was significantly associated with clinical progression at the last follow-up (hazard ratio, 5.1, P value, 0.04). Furthermore, the receipt of sRT correlated significantly with lower clinical progression at the last follow-up (hazard ratio, 0.2, P value, 0.03), whereas other clinical and tumor-specific parameters did not. Following surveillance-only and sRT, 29% (19 of 65) and 6% (2 of 36) of patients, respectively, showed clinical progression. In the sRT group, no significant difference was observed in FFP between patients who underwent sRT to the prostatic fossa versus those who received sRT to the prostatic fossa and pelvic lymph nodes, although the numbers in these groups were small. CONCLUSIONS This study suggests that salvage radiotherapy is associated with a decreased or delayed clinical progression in patients with biochemical recurrence following radical prostatectomy who have negative PSMA PET/CT scan results. The analysis also underscores the prognostic significance of the initial ISUP grade, with ISUP grade 5 being associated with worse outcomes. TRIAL REGISTRATION Registered September 14, 2016; NCT02899312 .
Collapse
Affiliation(s)
- Sara Harsini
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada
| | - Patrick Martineau
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Sonia Plaha
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada
| | - Heather Saprunoff
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada
| | - Catherine Chen
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada
| | - Julia Bishop
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada
| | - Scott Tyldesley
- Department of Radiation Oncology, BC Cancer, Vancouver, BC, Canada
| | - Don Wilson
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - François Bénard
- Department of Molecular Imaging and Therapy, BC Cancer Research Institute, 675 West 10th Ave, Vancouver, BC, Canada.
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
8
|
Kim M, Tamukong P, Galvan GC, Yang Q, De Hoedt A, Freeman MR, You S, Freedland S. Prostate cancers with distinct transcriptional programs in Black and White men. Genome Med 2024; 16:92. [PMID: 39044302 PMCID: PMC11267822 DOI: 10.1186/s13073-024-01361-0] [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: 12/27/2023] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Black men are at a higher risk of prostate cancer (PC) diagnosis and present with more high-grade PC than White men in an equal access setting. This study aimed to identify differential transcriptional regulation between Black and White men with PC. METHODS We performed microarray of radical prostatectomy tissue blocks from 305 Black and 238 White men treated at the Durham Veterans Affairs Medical Center. Differential expression, gene set enrichment analysis, master regulator analysis, and network modeling were conducted to compare gene expression by race. Findings were validated using external datasets that are available in the Gene Expression Omnibus (GEO) database. The first was a multi-institutional cohort of 1152 prostate cancer patients (596 Black, 556 White) with microarray data (GEO ID: GSE169038). The second was an Emory cohort of 106 patients (22 Black, 48 White, 36 men of unknown race) with RNA-seq data (GEO ID: GSE54460). Additionally, we analyzed androgen receptor (AR) chromatin binding profiles using paired AR ChIP-Seq datasets from Black and White men (GEO IDs: GSE18440 and GSE18441). RESULTS We identified 871 differentially expressed genes between Black and White men. White men had higher activity of MYC-related pathways, while Black men showed increased activity of inflammation, steroid hormone responses, and cancer progression-related pathways. We further identified the top 10 transcription factors (TFs) in Black patients, which formed a transcriptional regulatory network centered on the AR. The activities of this network and the pathways were significantly different in Black vs. White men across multiple cohorts and PC molecular subtypes. CONCLUSIONS These findings suggest PC in Black and White men have distinct tumor transcriptional profiles. Furthermore, a highly interactive TF network centered on AR drives differential gene expression in Black men. Additional study is needed to understand the degree to which these differences in transcriptional regulatory elements contribute to PC health disparities.
Collapse
Affiliation(s)
- Minhyung Kim
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Patrick Tamukong
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Qian Yang
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Michael R Freeman
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Stephen Freedland
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Veteran Affairs Health Care System, Durham, NC, USA.
| |
Collapse
|
9
|
Crawford ED, Bryce AH, Hussain MH, Agarwal N, Beltran H, Cooperberg MR, Petrylak DP, Shore N, Spratt DE, Tagawa ST, Antonarakis ES, Aparicio AM, Armstrong AJ, Boike TP, Calais J, Carducci MA, Chapin BF, Cookson MS, Davis JW, Dorff T, Eggener SE, Feng FY, Gleave M, Higano C, Iagaru A, Morgans AK, Morris M, Murray KS, Poage W, Rettig MB, Sartor O, Scher HI, Sieber P, Small E, Srinivas S, Yu EY, Zhang T, Koo PJ. Expert Perspectives on Controversies in Castration-Sensitive Prostate Cancer Management: Narrative Review and Report of the First US Prostate Cancer Conference Part 1. JU OPEN PLUS 2024; 2:e00029. [PMID: 38774466 PMCID: PMC11108024 DOI: 10.1097/ju9.0000000000000137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Purpose Castration-sensitive prostate cancer (CSPC) is a complex and heterogeneous condition encompassing a range of clinical presentations. As new approaches have expanded management options, clinicians are left with myriad questions and controversies regarding the optimal individualized management of CSPC. Materials and Methods The US Prostate Cancer Conference (USPCC) multidisciplinary panel was assembled to address the challenges of prostate cancer management. The first annual USPCC meeting included experts in urology, medical oncology, radiation oncology, and nuclear medicine. USPCC co-chairs and session moderators identified key areas of controversy and uncertainty in prostate cancer management and organized the sessions with multidisciplinary presentations and discussion. Throughout the meeting, experts responded to questions prepared by chairs and moderators to identify areas of agreement and controversy. Results The USPCC panel discussion and question responses for CSPC-related topics are presented. Key advances in CSPC management endorsed by USPCC experts included the development and clinical utilization of gene expression classifiers and artificial intelligence (AI) models for risk stratification and treatment selection in specific patient populations, the use of advanced imaging modalities in patients with clinically localized unfavorable intermediate or high-risk disease and those with biochemical recurrence, recommendations of doublet or triplet therapy for metastatic CSPC (mCSPC), and consideration of prostate and/or metastasis-directed radiation therapy in select patients with mCSPC. Conclusions CSPC is a diverse disease with many therapeutic options and the potential for adverse outcomes associated with either undertreatment or overtreatment. Future studies are needed to validate and clinically integrate novel technologies, including genomics, AI, and advanced imaging, to optimize outcomes among patients with CSPC.
Collapse
Affiliation(s)
- E. David Crawford
- Department of Urology, University of California San Diego, La Jolla, California
| | - Alan H. Bryce
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, Arizona
| | - Maha H. Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, Illinois
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Matthew R. Cooperberg
- Department of Urology, University of California at San Francisco, San Francisco, California
| | | | - Neal Shore
- Carolina Urologic Research Center/Genesis Care, Myrtle Beach, South Carolina
| | | | - Scott T. Tagawa
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, New York
| | | | - Ana M. Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew J. Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, North Carolina
| | | | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | | | - Brian F. Chapin
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael S. Cookson
- Department of Urology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma
| | - John W. Davis
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tanya Dorff
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Scott E. Eggener
- Departments of Surgery (Urology), University of Chicago Medical Center, Chicago, Illinois
| | - Felix Y. Feng
- Departments of Radiation Oncology, Urology, and Medicine, University of California San Francisco, San Francisco, California
| | - Martin Gleave
- Urological Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Celestia Higano
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Alicia K. Morgans
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Michael Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katie S. Murray
- Department of Urology, NYU Langone Health, New York, New York
| | - Wendy Poage
- Prostate Conditions Education Council, Centennial, Colorado
| | - Matthew B. Rettig
- Department of Medicine, Division of Hematology-Oncology, VA Greater Los Angeles, Los Angeles, California
- Departments of Medicine and Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Howard I. Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul Sieber
- Keystone Urology Specialists, Lancaster, Pennsylvania
| | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Sandy Srinivas
- Division of Medical Oncology, Stanford University Medical Center, Stanford, California
| | - Evan Y. Yu
- Department of Medicine, Division of Hematology & Oncology, University of Washington and Fred Hutchinson Cancer Center, Seattle, Washington
| | - Tian Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, Utah Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|
10
|
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
|
11
|
Baston C, Preda A, Iordache A, Olaru V, Surcel C, Sinescu I, Gingu C. How to Integrate Prostate Cancer Biomarkers in Urology Clinical Practice: An Update. Cancers (Basel) 2024; 16:316. [PMID: 38254807 PMCID: PMC10813985 DOI: 10.3390/cancers16020316] [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: 12/13/2023] [Revised: 01/04/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
Nowadays, the management of prostate cancer has become more and more challenging due to the increasing number of available treatment options, therapeutic agents, and our understanding of its carcinogenesis and disease progression. Moreover, currently available risk stratification systems used to facilitate clinical decision-making have limitations, particularly in providing a personalized and patient-centered management strategy. Although prognosis and prostate cancer-specific survival have improved in recent years, the heterogenous behavior of the disease among patients included in the same risk prognostic group negatively impacts not only our clinical decision-making but also oncological outcomes, irrespective of the treatment strategy. Several biomarkers, along with available tests, have been developed to help clinicians in difficult decision-making scenarios and guide management strategies. In this review article, we focus on the scientific evidence that supports the clinical use of several biomarkers considered by professional urological societies (and included in uro-oncological guidelines) in the diagnosis process and specific difficult management strategies for clinically localized or advanced prostate cancer.
Collapse
Affiliation(s)
- Catalin Baston
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Adrian Preda
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Alexandru Iordache
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Vlad Olaru
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Cristian Surcel
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Ioanel Sinescu
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| | - Constantin Gingu
- Department of Nephrology, Urology, Immunology and Immunology of Transplant, Dermatology, Allergology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.B.); (V.O.); (C.S.); (I.S.); (C.G.)
- Center of Uronephrology and Kidney Transplantation, Fundeni Clinical Institute, 258 Fundeni Street, 022328 Bucharest, Romania;
| |
Collapse
|
12
|
Boyer MJ, Carpenter DJ, Gingrich JR, Raman SR, Sirohi D, Tabriz AA, Rompre-Broduer A, Lunyera J, Basher F, Bitting RL, Kosinski A, Cantrell S, Gordon AM, Ear B, Gierisch JM, Jacobs M, Goldstein KM. Genomic classifiers and prognosis of localized prostate cancer: a systematic review. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-023-00766-z. [PMID: 38200096 DOI: 10.1038/s41391-023-00766-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/26/2023] [Accepted: 11/20/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Refinement of the risk classification for localized prostate cancer is warranted to aid in clinical decision making. A systematic analysis was undertaken to evaluate the prognostic ability of three genomic classifiers, Decipher, GPS, and Prolaris, for biochemical recurrence, development of metastases and prostate cancer-specific mortality in patients with localized prostate cancer. METHODS Data sources: MEDLINE, Embase, and Web of Science were queried for reports published from January 2010 to April 2022. STUDY SELECTION prospective or retrospective studies reporting prognosis for patients with localized prostate cancer. DATA EXTRACTION relevant data were extracted into a customized database by one researcher with a second overreading. Risk of bias was assessed using a validated tool for prognostic studies, Quality in Prognosis Studies (QUIPS). Disagreements were resolved by consensus or by input from a third reviewer. We assessed the certainty of evidence by GRADE incorporating adaptation for prognostic studies. RESULTS Data synthesis: a total of 39 studies (37 retrospective) involving over 10,000 patients were identified. Twenty-two assessed Decipher, 5 GPS, and 14 Prolaris. Thirty-four studies included patients who underwent prostatectomy. Based on very low to low certainty of evidence, each of the three genomic classifiers modestly improved upon the prognostic ability for biochemical recurrence, development of metastases, and prostate cancer-specific mortality compared to standard clinical risk-classification schemes. LIMITATIONS downgrading of confidence in the evidence stemmed largely from bias due to the retrospective nature of the studies, heterogeneity in treatment received, and era in which patients were treated (i.e., prior to the 2000s). CONCLUSIONS Genomic classifiers provide a small but consistent improvement upon the prognostic ability of clinical classification schemes, which may be helpful when treatment decisions are uncertain. However, evidence from current management-era data and of the predictive ability of these tests is needed.
Collapse
Affiliation(s)
- Matthew J Boyer
- Durham VA Health Care System, Durham, NC, USA.
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA.
| | | | - Jeffrey R Gingrich
- Durham VA Health Care System, Durham, NC, USA
- Department of Urology, Duke University School of Medicine, Durham, NC, USA
| | - Sudha R Raman
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Deepika Sirohi
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Amir Alishahi Tabriz
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Joseph Lunyera
- Division of General Internal Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Fahmin Basher
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Rhonda L Bitting
- Durham VA Health Care System, Durham, NC, USA
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Andrzej Kosinski
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Sarah Cantrell
- Duke University Medical Center Library & Archives, Duke University School of Medicine, Durham, NC, USA
| | | | - Belinda Ear
- Durham VA Health Care System, Durham, NC, USA
| | - Jennifer M Gierisch
- Durham VA Health Care System, Durham, NC, USA
- Division of General Internal Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Department of Population Health, Duke University School of Medicine, Durham, NC, USA
| | | | - Karen M Goldstein
- Durham VA Health Care System, Durham, NC, USA
- Division of General Internal Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
13
|
Naito Y, Kato M, Nagayama J, Sano Y, Matsuo K, Inoue S, Sano T, Ishida S, Matsukawa Y, Tsuzuki T, Akamatsu S. Recent insights on the clinical, pathological, and molecular features of intraductal carcinoma of the prostate. Int J Urol 2024; 31:7-16. [PMID: 37728330 DOI: 10.1111/iju.15299] [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: 06/26/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023]
Abstract
Intraductal carcinoma of the prostate, a unique histopathologic entity that is often observed (especially in advanced prostate cancer), is characterized by the proliferation of malignant cells within normal acini or ducts surrounded by a basement membrane. Intraductal carcinoma of the prostate is almost invariably associated with an adjacent high-grade carcinoma and is occasionally observed as an isolated subtype. Intraductal carcinoma of the prostate has been demonstrated to be an independent poor prognostic factor for all stages of cancer, whether localized, de novo metastatic, or castration-resistant. It also has a characteristic genetic profile, including high genomic instability. Recognizing and differentiating it from other pathologies is therefore important in patient management, and morphological diagnostic criteria for intraductal carcinoma of the prostate have been established. This review summarizes and outlines the clinical and pathological features, differential diagnosis, molecular aspects, and management of intraductal carcinoma of the prostate, as described in previous studies. We also present a discussion and future perspectives regarding intraductal carcinoma of the prostate.
Collapse
Affiliation(s)
- Yushi Naito
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masashi Kato
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Jun Nagayama
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuta Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuna Matsuo
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Satoshi Inoue
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tomoyasu Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shohei Ishida
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Aichi, Japan
| | - Shusuke Akamatsu
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
14
|
Adebahr S, Althaus A, Scharl S, Strouthos I, Farolfi A, Serani F, Lanzafame H, Trapp C, Koerber SA, Peeken JC, Vogel MME, Vrachimis A, Spohn SKB, Grosu AL, Kroeze SGC, Guckenberger M, Fanti S, Hruby G, Emmett L, Belka C, Schmidt-Hegemann NS, Henkenberens C, Aebersold DM, Wiegel T, Afshar-Oromieh A, Zamboglou C, Shelan M. The prognostic significance of a negative PSMA-PET scan prior to salvage radiotherapy following radical prostatectomy. Eur J Nucl Med Mol Imaging 2024; 51:558-567. [PMID: 37736808 PMCID: PMC10774185 DOI: 10.1007/s00259-023-06438-3] [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: 08/06/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Abstract
AIM The optimal management for early recurrent prostate cancer following radical prostatectomy (RP) in patients with negative prostate-specific membrane antigen positron-emission tomography (PSMA-PET) scan is an ongoing subject of debate. The aim of this study was to evaluate the outcome of salvage radiotherapy (SRT) in patients with biochemical recurrence with negative PSMA PET finding. METHODS This retrospective, multicenter (11 centers, 5 countries) analysis included patients who underwent SRT following biochemical recurrence (BR) of PC after RP without evidence of disease on PSMA-PET staging. Biochemical recurrence-free survival (bRFS), metastatic-free survival (MFS) and overall survival (OS) were assessed using Kaplan-Meier method. Multivariable Cox proportional hazards regression assessed predefined predictors of survival outcomes. RESULTS Three hundred patients were included, 253 (84.3%) received SRT to the prostate bed only, 46 (15.3%) additional elective pelvic nodal irradiation, respectively. Only 41 patients (13.7%) received concomitant androgen deprivation therapy (ADT). Median follow-up after SRT was 33 months (IQR: 20-46 months). Three-year bRFS, MFS, and OS following SRT were 73.9%, 87.8%, and 99.1%, respectively. Three-year bRFS was 77.5% and 48.3% for patients with PSA levels before PSMA-PET ≤ 0.5 ng/ml and > 0.5 ng/ml, respectively. Using univariate analysis, the International Society of Urological Pathology (ISUP) grade > 2 (p = 0.006), metastatic pelvic lymph nodes at surgery (p = 0.032), seminal vesicle involvement (p < 0.001), pre-SRT PSA level of > 0.5 ng/ml (p = 0.004), and lack of concomitant ADT (p = 0.023) were significantly associated with worse bRFS. On multivariate Cox proportional hazards, seminal vesicle infiltration (p = 0.007), ISUP score >2 (p = 0.048), and pre SRT PSA level > 0.5 ng/ml (p = 0.013) remained significantly associated with worse bRFS. CONCLUSION Favorable bRFS after SRT in patients with BR and negative PSMA-PET following RP was achieved. These data support the usage of early SRT for patients with negative PSMA-PET findings.
Collapse
Affiliation(s)
- Sonja Adebahr
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Freiburg, Germany
| | - Alexander Althaus
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Sophia Scharl
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Iosif Strouthos
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Nicosia, Cyprus
| | - Andrea Farolfi
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Serani
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Helena Lanzafame
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Christian Trapp
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Stefan A Koerber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Jan C Peeken
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Marco M E Vogel
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Alexis Vrachimis
- Department of Nuclear Medicine, German Oncology Center, University Hospital of the European University, Limassol, Cyprus
- C.A.R.I.C. Cancer Research & Innovation Center, Limassol, Cyprus
| | - Simon K B Spohn
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Stephanie G C Kroeze
- Radiation Oncology Center KSA-KSB, Canton Hospital of Aarau, Aarau, Switzerland
- Department of Radiation Oncology, University Hospital Zürich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zürich, University of Zurich, Zurich, Switzerland
| | - Stefano Fanti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - George Hruby
- Department of Radiation Oncology, Royal North Shore Hospital - University of Sydney, Sydney, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear medicine, St Vincent's Hospital Sydney, Sydney, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Nina-Sophie Schmidt-Hegemann
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Christoph Henkenberens
- Department of Radiotherapy and Special Oncology, Medical School Hannover, Hanover, Germany
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Freiburg, Germany
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Nicosia, Cyprus
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mohamed Shelan
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
15
|
Checcucci E, Oing C, Rescigno P. Genomic tests for persistence PSA after radical prostatectomy: smoke and mirrors or new effective tools for the clinical decision-making? Minerva Urol Nephrol 2023; 75:776-779. [PMID: 38126291 DOI: 10.23736/s2724-6051.23.05637-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Affiliation(s)
- Enrico Checcucci
- Department of Surgery, FPO-IRCCS Candiolo Cancer Institute, Candiolo, Turin, Italy -
| | - Christoph Oing
- Translational and Clinical Research Institute, Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Pasquale Rescigno
- Department of Surgery, FPO-IRCCS Candiolo Cancer Institute, Candiolo, Turin, Italy
- Translational and Clinical Research Institute, Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
16
|
Thomas M, Su YR, Rosenthal EA, Sakoda LC, Schmit SL, Timofeeva MN, Chen Z, Fernandez-Rozadilla C, Law PJ, Murphy N, Carreras-Torres R, Diez-Obrero V, van Duijnhoven FJB, Jiang S, Shin A, Wolk A, Phipps AI, Burnett-Hartman A, Gsur A, Chan AT, Zauber AG, Wu AH, Lindblom A, Um CY, Tangen CM, Gignoux C, Newton C, Haiman CA, Qu C, Bishop DT, Buchanan DD, Crosslin DR, Conti DV, Kim DH, Hauser E, White E, Siegel E, Schumacher FR, Rennert G, Giles GG, Hampel H, Brenner H, Oze I, Oh JH, Lee JK, Schneider JL, Chang-Claude J, Kim J, Huyghe JR, Zheng J, Hampe J, Greenson J, Hopper JL, Palmer JR, Visvanathan K, Matsuo K, Matsuda K, Jung KJ, Li L, Le Marchand L, Vodickova L, Bujanda L, Gunter MJ, Matejcic M, Jenkins MA, Slattery ML, D'Amato M, Wang M, Hoffmeister M, Woods MO, Kim M, Song M, Iwasaki M, Du M, Udaltsova N, Sawada N, Vodicka P, Campbell PT, Newcomb PA, Cai Q, Pearlman R, Pai RK, Schoen RE, Steinfelder RS, Haile RW, Vandenputtelaar R, Prentice RL, Küry S, Castellví-Bel S, Tsugane S, Berndt SI, Lee SC, Brezina S, Weinstein SJ, Chanock SJ, Jee SH, Kweon SS, Vadaparampil S, Harrison TA, Yamaji T, Keku TO, Vymetalkova V, Arndt V, Jia WH, Shu XO, Lin Y, Ahn YO, Stadler ZK, Van Guelpen B, Ulrich CM, Platz EA, Potter JD, Li CI, Meester R, Moreno V, Figueiredo JC, Casey G, Lansdorp Vogelaar I, Dunlop MG, Gruber SB, Hayes RB, Pharoah PDP, Houlston RS, Jarvik GP, Tomlinson IP, Zheng W, Corley DA, Peters U, Hsu L. Combining Asian and European genome-wide association studies of colorectal cancer improves risk prediction across racial and ethnic populations. Nat Commun 2023; 14:6147. [PMID: 37783704 PMCID: PMC10545678 DOI: 10.1038/s41467-023-41819-0] [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: 01/05/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
Polygenic risk scores (PRS) have great potential to guide precision colorectal cancer (CRC) prevention by identifying those at higher risk to undertake targeted screening. However, current PRS using European ancestry data have sub-optimal performance in non-European ancestry populations, limiting their utility among these populations. Towards addressing this deficiency, we expand PRS development for CRC by incorporating Asian ancestry data (21,731 cases; 47,444 controls) into European ancestry training datasets (78,473 cases; 107,143 controls). The AUC estimates (95% CI) of PRS are 0.63(0.62-0.64), 0.59(0.57-0.61), 0.62(0.60-0.63), and 0.65(0.63-0.66) in independent datasets including 1681-3651 cases and 8696-115,105 controls of Asian, Black/African American, Latinx/Hispanic, and non-Hispanic White, respectively. They are significantly better than the European-centric PRS in all four major US racial and ethnic groups (p-values < 0.05). Further inclusion of non-European ancestry populations, especially Black/African American and Latinx/Hispanic, is needed to improve the risk prediction and enhance equity in applying PRS in clinical practice.
Collapse
Affiliation(s)
- Minta Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Yu-Ru Su
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Biostatistics Division, Kaiser Permanente Washington Health Research Institute, Seattle, USA
| | - Elisabeth A Rosenthal
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, 98195, USA
| | - Lori C Sakoda
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
- Population and Cancer Prevention Program, Case Comprehensive Cancer Center, Cleveland, USA
| | - Maria N Timofeeva
- Danish Institute for Advanced Study (DIAS), Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, U, Germany
| | - Zhishan Chen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ceres Fernandez-Rozadilla
- Instituto de Investigacion Sanitaria de Santiago (IDIS), Choupana sn, 15706, Santiago de Compostela, Spain
- Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Reseach, London, SW7 3RP, UK
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Robert Carreras-Torres
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), Salt, 17190, Girona, Spain
| | - Virginia Diez-Obrero
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology, Barcelona, 08908, Spain
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute, Barcelona, 08908, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, 08908, Spain
| | | | - Shangqing Jiang
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Aesun Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Seoul, South Korea
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Amanda I Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Andrea Gsur
- .Center for Cancer Research, Medical University Vienna, Vienna, Austria
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Ann G Zauber
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna H Wu
- University of Southern California, Preventative Medicine, Los Angeles, CA, USA
| | - Annika Lindblom
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Y Um
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Catherine M Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chris Gignoux
- Colorado Center for Personalized Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Christina Newton
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - D Timothy Bishop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3000, Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3000, Australia
- Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, VIC, 3000, Australia
| | - David R Crosslin
- Department of Bioinformatics and Medical Education, University of Washington Medical Center, Seattle, WA, 98195, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dong-Hyun Kim
- Department of Social and Preventive Medicine, Hallym University College of Medicine, Okcheon-dong, South Korea
| | - Elizabeth Hauser
- VA Cooperative Studies Program Epidemiology Center, Durham Veterans Affairs Health Care System, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Erin Siegel
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Fredrick R Schumacher
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Gad Rennert
- Department of Community Medicine and Epidemiology, Lady Davis Carmel Medical Center, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Heather Hampel
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Isao Oze
- .Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Jae Hwan Oh
- .Research Institute and Hospital, National Cancer Center, Goyang, South Korea, South Korea
| | - Jeffrey K Lee
- .Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48104, USA
| | | | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Medical Centre Hamburg-Eppendorf, University Cancer Centre Hamburg (UCCH), Hamburg, Germany
| | - Jeongseon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Gyeonggi-do, South Korea
| | - Jeroen R Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jiayin Zheng
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jochen Hampe
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Joel Greenson
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48104, USA
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Epidemiology, School of Public Health and Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Julie R Palmer
- Slone Epidemiology Center, School of Medicine, Boston University, Boston, MA, USA
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Keitaro Matsuo
- Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Keum Ji Jung
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, VA, USA
| | | | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Luis Bujanda
- Department of Gastroenterology, Biodonostia Health Research Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | | | - Mark A Jenkins
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3000, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Mauro D'Amato
- Department of Medicine and Surgery, LUM University, Camassima, Italy
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, Derio, Spain
| | - Meilin Wang
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael O Woods
- Memorial University of Newfoundland, Discipline of Genetics, St. John's, Canada
| | - Michelle Kim
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Mingyang Song
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Departments of Epidemiology and Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Motoki Iwasaki
- Division of Epidemiology, National Cancer Center Institute for Cancer Control, National Cancer Center, Tokyo, Japan
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Mulong Du
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Natalia Udaltsova
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Norie Sawada
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Peter T Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel Pearlman
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Robert E Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Robert S Steinfelder
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Robert W Haile
- Samuel Oschin Comprehensive Cancer Institute, CEDARS-SINAI, Los Angeles, CA, USA
| | - Rosita Vandenputtelaar
- Department of Public Health, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ross L Prentice
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Sébastien Küry
- Nantes Université, CHU Nantes, Service de Génétique Médicale, F-44000, Nantes, France
| | - Sergi Castellví-Bel
- Gastroenterology Department, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Shoichiro Tsugane
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Soo Chin Lee
- National University Cancer Institute, Singapore, Singapore
| | - Stefanie Brezina
- .Center for Cancer Research, Medical University Vienna, Vienna, Austria
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Sun-Seog Kweon
- Department of Preventive Medicine, Chonnam National University Medical School, Gwangju, Korea
- Jeonnam Regional Cancer Center, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Susan Vadaparampil
- Departments of Epidemiology and Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Taiki Yamaji
- Division of Epidemiology, National Cancer Center Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Temitope O Keku
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| | - Veronika Vymetalkova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wei-Hua Jia
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Ou Shu
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Yoon-Ok Ahn
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Seoul, South Korea
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Cornelia M Ulrich
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Christopher I Li
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Reinier Meester
- Department of Public Health, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Victor Moreno
- Oncology Data Analytics Program, Catalan Institute of Oncology-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
- ONCOBEL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jane C Figueiredo
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Graham Casey
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Iris Lansdorp Vogelaar
- Department of Public Health, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Malcolm G Dunlop
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, U, Germany
| | - Stephen B Gruber
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Paul D P Pharoah
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Reseach, London, SW7 3RP, UK
| | - Gail P Jarvik
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, 98195, USA
| | - Ian P Tomlinson
- Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas A Corley
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
- Department of Gastroenterology, Kaiser Permanente Medical Center, San Francisco, CA, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
- Department of Biostatistics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Rafikova G, Gilyazova I, Enikeeva K, Pavlov V, Kzhyshkowska J. Prostate Cancer: Genetics, Epigenetics and the Need for Immunological Biomarkers. Int J Mol Sci 2023; 24:12797. [PMID: 37628978 PMCID: PMC10454494 DOI: 10.3390/ijms241612797] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Epidemiological data highlight prostate cancer as a significant global health issue, with high incidence and substantial impact on patients' quality of life. The prevalence of this disease is associated with various factors, including age, heredity, and race. Recent research in prostate cancer genetics has identified several genetic variants that may be associated with an increased risk of developing the disease. However, despite the significance of these findings, genetic markers for prostate cancer are not currently utilized in clinical practice as reliable indicators of the disease. In addition to genetics, epigenetic alterations also play a crucial role in prostate cancer development. Aberrant DNA methylation, changes in chromatin structure, and microRNA (miRNA) expression are major epigenetic events that influence oncogenesis. Existing markers for prostate cancer, such as prostate-specific antigen (PSA), have limitations in terms of sensitivity and specificity. The cost of testing, follow-up procedures, and treatment for false-positive results and overdiagnosis contributes to the overall healthcare expenditure. Improving the effectiveness of prostate cancer diagnosis and prognosis requires either narrowing the risk group by identifying new genetic factors or enhancing the sensitivity and specificity of existing markers. Immunological biomarkers (both circulating and intra-tumoral), including markers of immune response and immune dysfunction, represent a potentially useful area of research for enhancing the diagnosis and prognosis of prostate cancer. Our review emphasizes the need for developing novel immunological biomarkers to improve the diagnosis, prognosis, and management of prostate cancer. We highlight the most recent achievements in the identification of biomarkers provided by circulating monocytes and tumor-associated macrophages (TAMs). We highlight that monocyte-derived and TAM-derived biomarkers can enable to establish the missing links between genetic predisposition, hormonal metabolism and immune responses in prostate cancer.
Collapse
Affiliation(s)
- Guzel Rafikova
- Institute of Urology and Clinical Oncology, Bashkir State Medical University, 450077 Ufa, Russia (K.E.); (V.P.)
| | - Irina Gilyazova
- Institute of Urology and Clinical Oncology, Bashkir State Medical University, 450077 Ufa, Russia (K.E.); (V.P.)
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of the Russian Academy of Sciences, 450054 Ufa, Russia
| | - Kadriia Enikeeva
- Institute of Urology and Clinical Oncology, Bashkir State Medical University, 450077 Ufa, Russia (K.E.); (V.P.)
| | - Valentin Pavlov
- Institute of Urology and Clinical Oncology, Bashkir State Medical University, 450077 Ufa, Russia (K.E.); (V.P.)
| | - Julia Kzhyshkowska
- Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, 634050 Tomsk, Russia
- Genetic Technology Laboratory, Siberian State Medical University, 634050 Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, 68167 Mannheim, Germany
| |
Collapse
|
18
|
Bitting RL, Wu Y, Somarelli JA, Proudfoot JA, Liu Y, Davicioni E, George DJ, Armstrong AJ. Transcriptomic Signatures Associated With Outcomes in Recurrent Prostate Cancer Treated With Salvage Radiation, Androgen-Deprivation Therapy, and Enzalutamide: Correlative Analysis of the STREAM Trial. JCO Precis Oncol 2023; 7:e2300214. [PMID: 37595184 PMCID: PMC10581641 DOI: 10.1200/po.23.00214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/01/2023] [Accepted: 07/12/2023] [Indexed: 08/20/2023] Open
Abstract
PURPOSE Men with rising prostate-specific antigen (PSA) after radical prostatectomy (RP) may progress despite radiation and androgen-deprivation therapy (ADT). Tissue-based transcriptomic signatures can identify who may benefit from a more aggressive systemic approach. METHODS We performed a retrospective analysis of a prospective phase II multicenter trial of enzalutamide, ADT, and salvage radiotherapy in men with rising PSA after RP. Tumor tissue was analyzed using the Decipher platform for gene expression, including a novel prostate subtyping classifier, PTEN loss, homologous recombination deficiency (HRD), and ADT response. Cox models were used to associate signature scores with progression-free survival (PFS). RESULTS Of the 38 men enrolled, 31 had tissue with sufficient-quality RNA for genomic analysis. Luminal differentiated (LD) subtype tumors had the longest 3-year PFS at 89% compared with 19% in the luminal proliferating subtype. Men with signatures of PTEN loss (hazard ratio [HR], 1.32; 95% CI, 1.07 to 1.64; P = .01) or HRD (HR, 1.21; 95% CI, 1.05 to 1.39; P = .009) had worse PFS, while those with higher ADT response signature scores (HR, 0.75; 95% CI, 0.61 to 0.94; P = .01) were associated with improved PFS. Analysis of these signatures in a large cohort (n = 5,330) of RP samples from patients with biochemical recurrence found that these signatures provide complementary information related to outcomes with salvage radiation. CONCLUSION Despite aggressive systemic therapy with salvage radiation, nearly 50% of high-risk men relapse within 3 years. We show that LD and higher ADT sensitivity tumors had favorable outcomes. Those with a luminal proliferating subtype, PTEN loss, and/or HRD signatures had poor outcomes despite ADT/radiation and enzalutamide and may benefit from alternative approaches.
Collapse
Affiliation(s)
- Rhonda L. Bitting
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC
| | - Yuan Wu
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC
| | - Jason A. Somarelli
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC
| | | | - Yang Liu
- Veracyte, Inc, San Francisco, CA
| | | | - Daniel J. George
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC
| | | |
Collapse
|
19
|
Thomas M, Su YR, Rosenthal EA, Sakoda LC, Schmit SL, Timofeeva MN, Chen Z, Fernandez-Rozadilla C, Law PJ, Murphy N, Carreras-Torres R, Diez-Obrero V, van Duijnhoven FJ, Jiang S, Shin A, Wolk A, Phipps AI, Burnett-Hartman A, Gsur A, Chan AT, Zauber AG, Wu AH, Lindblom A, Um CY, Tangen CM, Gignoux C, Newton C, Haiman CA, Qu C, Bishop DT, Buchanan DD, Crosslin DR, Conti DV, Kim DH, Hauser E, White E, Siegel E, Schumacher FR, Rennert G, Giles GG, Hampel H, Brenner H, Oze I, Oh JH, Lee JK, Schneider JL, Chang-Claude J, Kim J, Huyghe JR, Zheng J, Hampe J, Greenson J, Hopper JL, Palmer JR, Visvanathan K, Matsuo K, Matsuda K, Jung KJ, Li L, Marchand LL, Vodickova L, Bujanda L, Gunter MJ, Matejcic M, Jenkins MA, Slattery ML, D'Amato M, Wang M, Hoffmeister M, Woods MO, Kim M, Song M, Iwasaki M, Du M, Udaltsova N, Sawada N, Vodicka P, Campbell PT, Newcomb PA, Cai Q, Pearlman R, Pai RK, Schoen RE, Steinfelder RS, Haile RW, Vandenputtelaar R, Prentice RL, Küry S, Castellví-Bel S, Tsugane S, Berndt SI, Lee SC, Brezina S, Weinstein SJ, Chanock SJ, Jee SH, Kweon SS, Vadaparampil S, Harrison TA, Yamaji T, Keku TO, Vymetalkova V, Arndt V, Jia WH, Shu XO, Lin Y, Ahn YO, Stadler ZK, Van Guelpen B, Ulrich CM, Platz EA, Potter JD, Li CI, Meester R, Moreno V, Figueiredo JC, Casey G, Vogelaar IL, Dunlop MG, Gruber SB, Hayes RB, Pharoah PDP, Houlston RS, Jarvik GP, Tomlinson IP, Zheng W, Corley DA, Peters U, Hsu L. Combining Asian-European Genome-Wide Association Studies of Colorectal Cancer Improves Risk Prediction Across Race and Ethnicity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.19.23284737. [PMID: 36789420 PMCID: PMC9928144 DOI: 10.1101/2023.01.19.23284737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Polygenic risk scores (PRS) have great potential to guide precision colorectal cancer (CRC) prevention by identifying those at higher risk to undertake targeted screening. However, current PRS using European ancestry data have sub-optimal performance in non-European ancestry populations, limiting their utility among these populations. Towards addressing this deficiency, we expanded PRS development for CRC by incorporating Asian ancestry data (21,731 cases; 47,444 controls) into European ancestry training datasets (78,473 cases; 107,143 controls). The AUC estimates (95% CI) of PRS were 0.63(0.62-0.64), 0.59(0.57-0.61), 0.62(0.60-0.63), and 0.65(0.63-0.66) in independent datasets including 1,681-3,651 cases and 8,696-115,105 controls of Asian, Black/African American, Latinx/Hispanic, and non-Hispanic White, respectively. They were significantly better than the European-centric PRS in all four major US racial and ethnic groups (p-values<0.05). Further inclusion of non-European ancestry populations, especially Black/African American and Latinx/Hispanic, is needed to improve the risk prediction and enhance equity in applying PRS in clinical practice.
Collapse
|
20
|
When to order genomic tests: development and external validation of a model to predict high-risk prostate cancer at the genotypic level. World J Urol 2023; 41:85-92. [PMID: 36484816 DOI: 10.1007/s00345-022-04240-8] [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: 07/16/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The aim of this study was to develop a model to predict high-genomic-risk prostate cancer (PCa) according to Decipher score, a validated 22 gene prognostic panel. By doing so, one might select the individuals who are likely to benefit from genomic testing and improve pre-op counseling about the need for adjuvant treatments. METHODS We retrospectively reviewed IRB-approved databases at two institutions. All patients had preoperative magnetic resonance imaging (MRI) and Decipher prostate radical prostatectomy (RP), a validated 22 gene prognostic panel. We used binary logistic regression to estimate high-risk Decipher (Decipher score > 0.60) probability on RP specimen. Area under the curve (AUC) and calibration were used to assess the accuracy of the model in the development and validation cohort. Decision curve analysis (DCA) was performed to assess the clinical benefit of the model. RESULTS The development and validation cohort included 622 and 185 patients with 283 (35%) and 80 (43%) of those with high-risk Decipher. The multivariable model included PSA density, biopsy Gleason Grade Group, percentage of positive cores and MRI extracapsular extension. AUC was 0.73 after leave-one-out cross-validation. DCA showed a clinical benefit in a range of probabilities between 15 and 60%. In the external validation cohort, AUC was 0.70 and calibration showed that the model underestimates the actual probability of the outcome. CONCLUSIONS The proposed model to predict high-risk Decipher score at RP is helpful to improve risk stratification of patients with PCa and to assess the need for additional testing and treatments.
Collapse
|
21
|
Spohn SKB, Draulans C, Kishan AU, Spratt D, Ross A, Maurer T, Tilki D, Berlin A, Blanchard P, Collins S, Bronsert P, Chen R, Pra AD, de Meerleer G, Eade T, Haustermans K, Hölscher T, Höcht S, Ghadjar P, Davicioni E, Heck M, Kerkmeijer LGW, Kirste S, Tselis N, Tran PT, Pinkawa M, Pommier P, Deltas C, Schmidt-Hegemann NS, Wiegel T, Zilli T, Tree AC, Qiu X, Murthy V, Epstein JI, Graztke C, Gao X, Grosu AL, Kamran SC, Zamboglou C. Genomic Classifiers in Personalized Prostate Cancer Radiation Therapy Approaches: A Systematic Review and Future Perspectives Based on International Consensus. Int J Radiat Oncol Biol Phys 2022:S0360-3016(22)03691-4. [PMID: 36596346 DOI: 10.1016/j.ijrobp.2022.12.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/09/2022] [Accepted: 12/24/2022] [Indexed: 01/01/2023]
Abstract
Current risk-stratification systems for prostate cancer (PCa) do not sufficiently reflect the disease heterogeneity. Genomic classifiers (GC) enable improved risk stratification after surgery, but less data exist for patients treated with definitive radiation therapy (RT) or RT in oligo-/metastatic disease stages. To guide future perspectives of GCs for RT, we conducted (1) a systematic review on the evidence of GCs for patients treated with RT and (2) a survey of experts using the Delphi method, addressing the role of GCs in personalized treatments to identify relevant fields of future clinical and translational research. We performed a systematic review and screened ongoing clinical trials on ClinicalTrials.gov. Based on these results, a multidisciplinary international team of experts received an adapted Delphi method survey. Thirty-one and 30 experts answered round 1 and round 2, respectively. Questions with ≥75% agreement were considered relevant and included in the qualitative synthesis. Evidence for GCs as predictive biomarkers is mainly available to the postoperative RT setting. Validation of GCs as prognostic markers in the definitive RT setting is emerging. Experts used GCs in patients with PCa with extensive metastases (30%), in postoperative settings (27%), and in newly diagnosed PCa (23%). Forty-seven percent of experts do not currently use GCs in clinical practice. Expert consensus demonstrates that GCs are promising tools to improve risk-stratification in primary and oligo-/metastatic patients in addition to existing classifications. Experts were convinced that GCs might guide treatment decisions in terms of RT-field definition and intensification/deintensification in various disease stages. This work confirms the value of GCs and the promising evidence of GC utility in the setting of RT. Additional studies of GCs as prognostic biomarkers are anticipated and form the basis for future studies addressing predictive capabilities of GCs to optimize RT and systemic therapy. The expert consensus points out future directions for GC research in the management of PCa.
Collapse
Affiliation(s)
- Simon K B Spohn
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Cédric Draulans
- Department of Radiation Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU Leuven, Belgium
| | - Amar U Kishan
- Departments of Radiation Oncology and Urology, University of California, Los Angeles, California
| | - Daniel Spratt
- Department of Radiation Oncology, UH Seidman Cancer Center, Case Western Reserve University
| | - Ashley Ross
- Department of Urology, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Alejandro Berlin
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, and Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network. Toronto, Canada
| | - Pierre Blanchard
- Department of Radiation Oncology, Gustave Roussy, Oncostat U1018, Inserm, Paris-Saclay University, Villejuif, France
| | - Sean Collins
- Department of Radiation Medicine, Medstar Georgetown University Hospital, Washington, DC
| | - Peter Bronsert
- Institute for Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ronald Chen
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, Kansas
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami, Miller School of Medicine
| | - Gert de Meerleer
- Department of Radiation Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU Leuven, Belgium
| | - Thomas Eade
- Northern Sydney Cancer Centre, Radiation Oncology Unit, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Karin Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU Leuven, Belgium
| | - Tobias Hölscher
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan Höcht
- Xcare Practices Dept. Radiotherapy, Saarlouis, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin
| | | | - Matthias Heck
- Department of Urology, Rechts der Isar Medical Center, Technical University of Munich, Germany
| | - Linda G W Kerkmeijer
- Department of Radiation Oncology, Radboud University Medical Center, The Netherlands
| | - Simon Kirste
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Nikolaos Tselis
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland
| | - Michael Pinkawa
- Department of Radiation Oncology, MediClin Robert Janker Klinik Bonn, Germany
| | - Pascal Pommier
- Department of Radiation Oncology, Centre Léon Bérard, Lyon, France
| | - Constantinos Deltas
- Molecular Medicine Research Center and Laboratory of Molecular and Medical Genetics, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | | | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Thomas Zilli
- Department of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Alison C Tree
- Department of Radiotherapy, Royal Marsden Hospital and the Institute of Cancer Research, London, United Kingdom
| | - Xuefeng Qiu
- Department of Urology, Medical School of Nanjing University, Affiliated Drum Tower Hospital, Nanjing, China
| | - Vedang Murthy
- Department of Radiation Oncology, ACTREC, Tata Memorial Centre, Homi Bhabha National University, India
| | - Jonathan I Epstein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christian Graztke
- Department of Urology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xin Gao
- Department of Internal Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Anca L Grosu
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Constantinos Zamboglou
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Oncology Center, European University of Cyprus, Limassol, Cyprus
| |
Collapse
|
22
|
The CAPRA&PDE4D5/7/9 Prognostic Model Is Significantly Associated with Adverse Post-Surgical Pathology Outcomes. Cancers (Basel) 2022; 15:cancers15010262. [PMID: 36612262 PMCID: PMC9818961 DOI: 10.3390/cancers15010262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023] Open
Abstract
Objectives: To investigate the association of the prognostic risk score CAPRA&PDE4D5/7/9 as measured on pre-surgical diagnostic needle biopsy tissue with pathological outcomes after radical prostatectomies in a clinically low−intermediate-risk patient cohort. Patients and Methods: RNA was extracted from biopsy punches of diagnostic needle biopsies. The patient cohort comprises n = 151 patients; of those n = 84 had low−intermediate clinical risk based on the CAPRA score and DRE clinical stage <cT3. This cohort (n = 84) was investigated for pathology outcomes in this study. RT-qPCR was performed to determine PDE4D5, PDE4D7 and PDE4D9 transcript scores in the cohorts. The CAPRA score was inferred from the relevant clinical data (patient age, PSA, cT, biopsy Gleason, and percentage tumor positive biopsy cores). Logistic regression was used to combine the PDE4D5, PDE4D7 and PDE4D9 scores to build a PDE4D5/7/9_BCR regression model. The CAPRA&PDE4D5/7/9_BCR risk score used was same as previously published. Results: We investigated three post-surgical outcomes in this study: (i) Adverse Pathology (any ISUP pathological Gleason grade >2, or pathological pT stage > pT3a, or tumor penetrated prostate capsular status, or pN1 disease); (ii) any ISUP pathological Gleason >2; (iii) any ISUP pathological Gleason >1. In the n = 84 patients with low to intermediate clinical risk profiles, the clinical-genomics CAPRA&PDE4D5/7/9_BCR risk score was significantly lower in patients with favorable vs. unfavorable outcomes. In univariable logistic regression modeling the genomics PDE4D5/7/9_BCR as well as the clinical-genomics CAPRA&PDE4D5/7/9_BCR combination model were significantly associated with all three post-surgical pathology outcomes (p = 0.02, p = 0.0004, p = 0.04; and p = 0.01, p = 0.0002, p = 0.01, respectively). The clinically used PRIAS criteria for the selection of low-risk candidate patients for active surveillance (AS) were not significantly associated with any of the three tested post-operative pathology outcomes (p = 0.3, p = 0.1, p = 0.1, respectively). In multivariable analysis adjusted for the CAPRA score, the genomics PDE4D5/7/9_BCR risk score remained significant for the outcomes of adverse pathology (p = 0.04) and ISUP pathological Gleason >2 (p = 0.004). The negative predictive value of the CAPRA&PDE4D5/7/9_BCR risk score using the low-risk cut-off (0.1) for the three pathological endpoints was 82.0%, 100%, and 59.1%, respectively for a selected low-risk cohort of n = 22 patients (26.2% of the entire cohort) compared to 72.1%, 94.4%, and 55.6% for n = 18 low-risk patients (21.4% of the total cohort) selected based on the PRIAS inclusion criteria. Conclusion: In this study, we have shown that the previously reported clinical-genomics prostate cancer risk model CAPRA&PDE4D5/7/9_BCR which was developed to predict biological outcomes after surgery of primary prostate cancer is also significantly associated with post-surgical pathology outcomes. The risk score predicts adverse pathology independent of the clinical risk metrics. Compared to clinically used active surveillance inclusion criteria, the clinical-genomics CAPRA&PDE4D5/7/9_BCR risk model selects 22% (n = 8) more low-risk patients with higher negative predictive value to experience unfavorable post-operative pathology outcomes.
Collapse
|
23
|
Lone Z, Benidir T, Rainey M, Nair M, Davicioni E, Gibb EA, Williamson S, Gupta S, Chaim Ornstein M, Tendulkar R, Weight C, Nguyen JK, Klein EA, Mian OY. Transcriptomic Features of Cribriform and Intraductal Carcinoma of the Prostate. Eur Urol Focus 2022; 8:1575-1582. [PMID: 35662504 DOI: 10.1016/j.euf.2022.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/28/2022] [Accepted: 05/22/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Cribriform (CF) and/or intraductal carcinoma (IDC) are associated with more aggressive prostate cancer (CaP) and worse outcomes. OBJECTIVE The transcriptomic features that typify CF/IDC are not well described and the capacity for clinically utilized genomic classifiers to improve risk modeling for CF/IDC remains undefined. DESIGN, SETTING, AND PARTICIPANTS We performed a retrospective review of CaP patients who had Decipher testing at a single high-volume institution. Index lesions from radical prostatectomy specimens were identified by genitourinary pathologists who simultaneously reviewed prostatectomy specimens for the presence of CF and IDC features. Patients were grouped based on pathologic features, specifically the absence of CF/IDC (CF-/IDC-), CF positive only (CF+/IDC-), and CF/IDC positive (CF+/IDC+). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Clinical, pathologic, and genomic categorical variables were assessed using the Pearson chi-square test, while quantitative variables were assessed with the Kruskal-Wallis test. Multivariable logistic regression was used to identify the predictors of high-risk Decipher scores (>0.60). A gene set enrichment analysis was performed to identify genes and gene networks associated with CF/IDC status. RESULTS AND LIMITATIONS A total of 463 patients were included. Patients who were CF+/IDC+ had the highest Decipher risk scores (CF+/IDC+: 0.79 vs CF+/IDC-: 0.71 vs CF-/IDC-: 0.56, p < 0.001). On multivariate logistic regression, predictors of high-risk Decipher scores included the presence of CF, both alone (CF+/IDC-; odds ratio [OR]: 5.45, p < 0.001) or in combination with positive IDC status (CF+/IDC+; OR: 6.87, p < 0.001). On the gene set enrichment analysis, MYC pathway upregulation was significantly enriched in tumor samples from CF/IDC-positive patients (normalized enrichment score [NES]: 1.65, p = 0.046). Other enriched pathways included E2F targets (NES: 1.69, p = 0.031) and oxidative phosphorylation (NES: 1.68, =0 .033). CONCLUSIONS This is the largest series identifying an association between a clinically validated genomic classifier and the presence of CF and IDC at radical prostatectomy. Tumors with CF and intraductal features were associated with aggressive transcriptomic signatures. PATIENT SUMMARY Genomic-based tests are becoming readily available for the management of prostate cancer. We observed that Decipher, a commonly used genomic test in prostate cancer, correlates with unfavorable features in tissue specimens.
Collapse
Affiliation(s)
- Zaeem Lone
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA.
| | - Tarik Benidir
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Monica Nair
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | | | | | - Sean Williamson
- Cleveland Clinic Department of Pathology, Cleveland, OH, USA
| | - Shilpa Gupta
- Cleveland Clinic Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | | | - Rahul Tendulkar
- Cleveland Clinic Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Christopher Weight
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jane K Nguyen
- Cleveland Clinic Department of Pathology, Cleveland, OH, USA
| | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Omar Y Mian
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA; Cleveland Clinic Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
24
|
Bogdanova NV, Radmanesh H, Ramachandran D, Knoechelmann AC, Christiansen H, Derlin T, von Klot CAJ, Merten R, Henkenberens C. The Prognostic Value of Liquid Biopsies for Benefit of Salvage Radiotherapy in Relapsed Oligometastatic Prostate Cancer. Cancers (Basel) 2022; 14:cancers14174095. [PMID: 36077632 PMCID: PMC9454496 DOI: 10.3390/cancers14174095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Simple Summary Around 30% of patients with oligometastatic prostate cancer relapse will benefit from local PET/CT-guided ablative radiotherapy (RT) with improved progression-free and ADT (Androgene Deprivation Therapy)-free survivals. Therefore, there is an urgent need for predictive testing for therapeutic benefits prior to initiation. Various tests have already been established on tumor specimens for the prediction of prostate cancer’s behavior or therapy outcome. However, in imaging-proven relapse tumor tissue from the local recurrence or metastases is often not available. Hence, there is a need for a liquid biopsy-based testing. We aimed to assess the prognostic value of CTCs- associated mRNA and blood-derived RNA for the benefit of PSMA PET-guided salvage RT in oligometastatic prostate cancer relapses. Significant correlations were found between the relative transcript levels of several investigated genes and clinicopathological parameters. Furthermore, distinct “transcriptional signatures” were found in patients with temporary and long-term benefits from RT. Abstract To assess the prognostic value of “liquid biopsies” for the benefit of salvage RT in oligometastatic prostate cancer relapse, we enrolled 44 patients in the study between the years 2016 and 2020. All the patients were diagnosed as having an oligometastatic prostate cancer relapse on prostate-specific membrane antigen (PSMA)-targeted PET-CT and underwent irradiation at the Department of Radiotherapy at the Hannover Medical School. Tumor cells and total RNA, enriched from the liquid biopsies of patients, were processed for the subsequent quantification analysis of relative transcript levels in real-time PCR. In total, 54 gene transcripts known or suggested to be associated with prostate cancer or treatment outcome were prioritized for analysis. We found significant correlations between the relative transcript levels of several investigated genes and the Gleason score, PSA (prostate-specific antigen) value, or UICC stage (tumor node metastasis -TNM classification of malignant tumors from Union for International Cancer Control). Furthermore, a significant association of MTCO2, FOXM1, SREBF1, HOXB7, FDXR, and MTRNR transcript profiles was found with a temporary and/or long-term benefit from RT. Further studies on larger patients cohorts are necessary to prove our preliminary findings for establishing liquid biopsy tests as a predictive examination method prior to salvage RT.
Collapse
Affiliation(s)
- Natalia V. Bogdanova
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Hoda Radmanesh
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Dhanya Ramachandran
- Gynecology Research Unit, Clinics of Obstetrics and Gynaecology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Hans Christiansen
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | | | - Roland Merten
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence: ; Tel.: +49-(0)-511-532-3590
| | | |
Collapse
|
25
|
Chiu PKF, Lee EKC, Chan MTY, Chan WHC, Cheung MH, Lam MHC, Ma ESK, Poon DMC. Genetic Testing and Its Clinical Application in Prostate Cancer Management: Consensus Statements from the Hong Kong Urological Association and Hong Kong Society of Uro-Oncology. Front Oncol 2022; 12:962958. [PMID: 35924163 PMCID: PMC9339641 DOI: 10.3389/fonc.2022.962958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Background In recent years, indications for genetic testing in prostate cancer (PC) have expanded from patients with a family history of prostate and/or related cancers to those with advanced castration-resistant disease, and even to early PC patients for determination of the appropriateness of active surveillance. The current consensus aims to provide guidance to urologists, oncologists and pathologists working with Asian PC patients on who and what to test for in selected populations. Methods A joint consensus panel from the Hong Kong Urological Association and Hong Kong Society of Uro-Oncology was convened over a series of 5 physical and virtual meetings. A background literature search on genetic testing in PC was performed in PubMed, ClinicalKey, EBSCOHost, Ovid and ProQuest, and three working subgroups were formed to review and present the relevant evidence. Meeting agendas adopted a modified Delphi approach to ensure that discussions proceed in a structured, iterative and balanced manner, which was followed by an anonymous voting on candidate statements. Of 5 available answer options, a consensus statement was accepted if ≥ 75% of the panelists chose “Accept Completely” (Option A) or “Accept with Some Reservation” (Option B). Results The consensus was structured into three parts: indications for testing, testing methods, and therapeutic implications. A list of 35 candidate statements were developed, of which 31 were accepted. The statements addressed questions on the application of PC genetic testing data and guidelines to Asian patients, including patient selection for germline testing, selection of gene panel and tissue sample, provision of genetic counseling, and use of novel systemic treatments in metastatic castration-resistant PC patients. Conclusion This consensus provides guidance to urologists, oncologists and pathologists working with Asian patients on indications for genetic testing, testing methods and technical considerations, and associated therapeutic implications.
Collapse
Affiliation(s)
- Peter K. F. Chiu
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eric K. C. Lee
- Department of Clinical Oncology, Tuen Mun Hospital, Hong Kong SAR, China
| | - Marco T. Y. Chan
- Division of Urology, Department of Surgery, Tuen Mun Hospital, Hong Kong SAR, China
| | - Wilson H. C. Chan
- Division of Urology, Department of Surgery, United Christian Hospital, Hong Kong SAR, China
| | - M. H. Cheung
- Division of Urology, Department of Surgery, Tseung Kwan O Hospital, Hong Kong SAR, China
| | - Martin H. C. Lam
- Department of Oncology, United Christian Hospital, Hong Kong SAR, China
| | - Edmond S. K. Ma
- Department of Pathology, Hong Kong Sanatorium and Hospital, Hong Kong SAR, China
| | - Darren M. C. Poon
- Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- Comprehensive Oncology Centre, Hong Kong Sanatorium and Hospital, Hong Kong SAR, China
- *Correspondence: Darren M. C. Poon,
| |
Collapse
|
26
|
Cao Y, Deville C. Adjuvant Radiation for Pathologically Node-Positive Prostate Cancer: Evidence When Early Salvage May Not Be Early Enough. J Clin Oncol 2022; 40:2179-2182. [PMID: 35605175 DOI: 10.1200/jco.22.00645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yilin Cao
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Curtiland Deville
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
27
|
Pantazopoulos H, Diop MK, Grosset AA, Rouleau-Gagné F, Al-Saleh A, Boblea T, Trudel D. Intraductal Carcinoma of the Prostate as a Cause of Prostate Cancer Metastasis: A Molecular Portrait. Cancers (Basel) 2022; 14:820. [PMID: 35159086 PMCID: PMC8834356 DOI: 10.3390/cancers14030820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
Intraductal carcinoma of the prostate (IDC-P) is one of the most aggressive types of prostate cancer (PCa). IDC-P is identified in approximately 20% of PCa patients and is associated with recurrence, metastasis, and PCa-specific death. The main feature of this histological variant is the colonization of benign glands by PCa cells. Although IDC-P is a well-recognized independent parameter for metastasis, mechanisms by which IDC-P cells can spread and colonize other tissues are not fully known. In this review, we discuss the molecular portraits of IDC-P determined by immunohistochemistry and genomic approaches and highlight the areas in which more research is needed.
Collapse
Affiliation(s)
- Helen Pantazopoulos
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Mame-Kany Diop
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Andrée-Anne Grosset
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Frédérique Rouleau-Gagné
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Afnan Al-Saleh
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Teodora Boblea
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
| | - Dominique Trudel
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Department of Pathology, Centre Hospitalier de l’Université de Montréal (CHUM), 1051 Sanguinet, Montreal, QC H2X 0C1, Canada
| |
Collapse
|
28
|
Prognostic Genomic Tissue-Based Biomarkers in the Treatment of Localized Prostate Cancer. J Pers Med 2022; 12:jpm12010065. [PMID: 35055380 PMCID: PMC8781984 DOI: 10.3390/jpm12010065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/15/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
In localized prostate cancer clinicopathologic variables have been used to develop prognostic nomograms quantifying the probability of locally advanced disease, of pelvic lymph node and distant metastasis at diagnosis or the probability of recurrence after radical treatment of the primary tumor. These tools although essential in daily clinical practice for the management of such a heterogeneous disease, which can be cured with a wide spectrum of treatment strategies (i.e., active surveillance, RP and radiation therapy), do not allow the precise distinction of an indolent instead of an aggressive disease. In recent years, several prognostic biomarkers have been tested, combined with the currently available clinicopathologic prognostic tools, in order to improve the decision-making process. In the following article, we reviewed the literature of the last 10 years and gave an overview report on commercially available tissue-based biomarkers and more specifically on mRNA-based gene expression classifiers. To date, these genomic tests have been widely investigated, demonstrating rigorous quality criteria including reproducibility, linearity, analytical accuracy, precision, and a positive impact in the clinical decision-making process. Albeit data published in literature, the systematic use of these tests in prostate cancer is currently not recommended due to insufficient evidence.
Collapse
|
29
|
Farha MW, Salami SS. Biomarkers for prostate cancer detection and risk stratification. Ther Adv Urol 2022; 14:17562872221103988. [PMID: 35719272 PMCID: PMC9201356 DOI: 10.1177/17562872221103988] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
Although prostate cancer (PCa) is the most commonly diagnosed cancer in men, most patients do not die from the disease. Prostate specific antigen (PSA), the most widely used oncologic biomarker, has revolutionized screening and early detection, resulting in reduced proportion of patients presenting with advanced disease. However, given the inherent limitations of PSA, additional diagnostic and prognostic tools are needed to facilitate early detection and accurate risk stratification of disease. Serum, urine, and tissue-based biomarkers are increasingly being incorporated into the clinical care paradigm, but there is still a limited understanding of how to use them most effectively. In the current article, we review test characteristics and clinical performance data for both serum [4 K score, prostate health index (phi)] and urine [SelectMDx, ExoDx Prostate Intelliscore, MyProstateScore (MPS), and PCa antigen 3 (PCA3)] biomarkers to aid decisions regarding initial or repeat biopsies as well as tissue-based biomarkers (Confirm MDx, Decipher, Oncotype Dx, and Polaris) aimed at risk stratifying patients and identifying those patients most likely to benefit from treatment versus surveillance or monotherapy versus multi-modal therapy.
Collapse
Affiliation(s)
- Mark W. Farha
- University of Michigan Medical School, Ann
Arbor, MI, USA
| | - Simpa S. Salami
- Department of Urology, Michigan Medicine, 1500
E. Medical Center Dr., 7306 Rogel Cancer Center, Ann Arbor, MI 48109-5948,
USA
- University of Michigan Medical School, Ann
Arbor, MI, USA
- Rogel Cancer Center, University of Michigan,
Ann Arbor, MI, USA
| |
Collapse
|
30
|
Elbanna M, Chowdhury NN, Rhome R, Fishel ML. Clinical and Preclinical Outcomes of Combining Targeted Therapy With Radiotherapy. Front Oncol 2021; 11:749496. [PMID: 34733787 PMCID: PMC8558533 DOI: 10.3389/fonc.2021.749496] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022] Open
Abstract
In the era of precision medicine, radiation medicine is currently focused on the precise delivery of highly conformal radiation treatments. However, the tremendous developments in targeted therapy are yet to fulfill their full promise and arguably have the potential to dramatically enhance the radiation therapeutic ratio. The increased ability to molecularly profile tumors both at diagnosis and at relapse and the co-incident progress in the field of radiogenomics could potentially pave the way for a more personalized approach to radiation treatment in contrast to the current ‘‘one size fits all’’ paradigm. Few clinical trials to date have shown an improved clinical outcome when combining targeted agents with radiation therapy, however, most have failed to show benefit, which is arguably due to limited preclinical data. Several key molecular pathways could theoretically enhance therapeutic effect of radiation when rationally targeted either by directly enhancing tumor cell kill or indirectly through the abscopal effect of radiation when combined with novel immunotherapies. The timing of combining molecular targeted therapy with radiation is also important to determine and could greatly affect the outcome depending on which pathway is being inhibited.
Collapse
Affiliation(s)
- May Elbanna
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Nayela N Chowdhury
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ryan Rhome
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Melissa L Fishel
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
31
|
Development of a MicroRNA Signature Predictive of Recurrence and Survival in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13205168. [PMID: 34680317 PMCID: PMC8534163 DOI: 10.3390/cancers13205168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Optimal patient selection for radiotherapy in pancreatic cancer is unestablished and may be improved with molecular profiling. To this end, we developed and validated a microRNA signature that predicted for worse locoregional recurrence and overall survival in patients with resectable pancreatic cancer. In a separate cohort of patients with borderline resectable and locally advanced pancreatic cancer, this risk signature was also predictive of worse locoregional recurrence, distant recurrence, and overall survival. Additionally, borderline resectable or locally advanced patients who had high risk score and did not receive radiation had worse outcomes compared to patients who either had low risk score or received radiation, irrespective of risk score. This risk signature may be useful in assessing patient prognosis and tailor therapy in patients with resectable, borderline resectable, or locally advanced pancreatic cancer, but requires further study. Abstract Background: Optimal patient selection for radiotherapy in pancreatic ductal adenocarcinoma (PDAC) is unestablished. Molecular profiling may select patients at high risk for locoregional recurrence (LRR) who would benefit from radiation. Methods: We included resectable pancreatic cancer (R-PDAC) patients, divided into training and validation cohorts, treated among three institutions with surgery and adjuvant chemotherapy, and borderline resectable or locally advanced pancreatic cancer (BR/LA-PDAC) patients treated with chemotherapy with or without radiation at the primary study institution. We isolated RNA from R-PDAC surgical specimens. Using NanoString, we identified miRNAs differentially expressed between normal and malignant pancreatic tissue. ElasticNet regression identified two miRNAs most predictive of LRR in the training cohort, miR-181b/d and miR-575, which were used to generate a risk score (RS). We evaluated the association of the median-dichotomized RS with recurrence and overall survival (OS). Results: We identified 183 R-PDAC and 77 BR/LA-PDAC patients with median follow up of 37 months treated between 2001 and 2014. On multivariable analysis of the R-PDAC training cohort (n = 90), RS was associated with worse LRR (HR = 1.34; 95%CI 1.27–11.38; p = 0.017) and OS (HR = 2.89; 95%CI 1.10–4.76; p = 0.027). In the R-PDAC validation cohort, RS was associated with worse LRR (HR = 2.39; 95%CI 1.03–5.54; p = 0.042), but not OS (p = 0.087). For BR/LA-PDAC, RS was associated with worse LRR (HR = 2.71; 95%CI 1.14–6.48; p = 0.025), DR (HR = 1.93; 95%CI 1.10–3.38; p = 0.022), and OS (HR = 1.97; 95%CI 1.17–3.34; p = 0.011). Additionally, after stratifying by RS and receipt of radiation in BR/LA-PDAC patients, high RS patients who did not receive radiation had worse LRR (p = 0.018), DR (p = 0.006), and OS (p < 0.001) compared to patients with either low RS or patients who received radiation, irrespective of RS. Conclusions: RS predicted worse LRR and OS in R-PDAC and worse LRR, DR, and OS in BR/LA-PDAC. This may select patients who would benefit from radiation and should be validated prospectively.
Collapse
|
32
|
Meehan J, Gray M, Martínez-Pérez C, Kay C, McLaren D, Turnbull AK. Tissue- and Liquid-Based Biomarkers in Prostate Cancer Precision Medicine. J Pers Med 2021; 11:jpm11070664. [PMID: 34357131 PMCID: PMC8306523 DOI: 10.3390/jpm11070664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022] Open
Abstract
Worldwide, prostate cancer (PC) is the second-most-frequently diagnosed male cancer and the fifth-most-common cause of all cancer-related deaths. Suspicion of PC in a patient is largely based upon clinical signs and the use of prostate-specific antigen (PSA) levels. Although PSA levels have been criticised for a lack of specificity, leading to PC over-diagnosis, it is still the most commonly used biomarker in PC management. Unfortunately, PC is extremely heterogeneous, and it can be difficult to stratify patients whose tumours are unlikely to progress from those that are aggressive and require treatment intensification. Although PC-specific biomarker research has previously focused on disease diagnosis, there is an unmet clinical need for novel prognostic, predictive and treatment response biomarkers that can be used to provide a precision medicine approach to PC management. In particular, the identification of biomarkers at the time of screening/diagnosis that can provide an indication of disease aggressiveness is perhaps the greatest current unmet clinical need in PC management. Largely through advances in genomic and proteomic techniques, exciting pre-clinical and clinical research is continuing to identify potential tissue, blood and urine-based PC-specific biomarkers that may in the future supplement or replace current standard practices. In this review, we describe how PC-specific biomarker research is progressing, including the evolution of PSA-based tests and those novel assays that have gained clinical approval. We also describe alternative diagnostic biomarkers to PSA, in addition to biomarkers that can predict PC aggressiveness and biomarkers that can predict response to certain therapies. We believe that novel biomarker research has the potential to make significant improvements to the clinical management of this disease in the near future.
Collapse
Affiliation(s)
- James Meehan
- Translational Oncology Research Group, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.M.-P.); (C.K.); (A.K.T.)
- Correspondence:
| | - Mark Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, UK;
| | - Carlos Martínez-Pérez
- Translational Oncology Research Group, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.M.-P.); (C.K.); (A.K.T.)
- Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Charlene Kay
- Translational Oncology Research Group, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.M.-P.); (C.K.); (A.K.T.)
- Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Duncan McLaren
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh EH4 2XU, UK;
| | - Arran K. Turnbull
- Translational Oncology Research Group, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.M.-P.); (C.K.); (A.K.T.)
- Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
33
|
Corradi JP, Cumarasamy CW, Staff I, Tortora J, Salner A, McLaughlin T, Wagner J. Identification of a five gene signature to predict time to biochemical recurrence after radical prostatectomy. Prostate 2021; 81:694-702. [PMID: 34002865 DOI: 10.1002/pros.24150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/01/2021] [Accepted: 04/26/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Identification of novel biomarkers associated with high-risk prostate cancer or biochemical recurrence can drive improvement in detection, prognosis, and treatment. However, studies can be limited by small sample sizes and sparse clinical follow-up data. We utilized a large sample of prostate specimens to identify a predictive model of biochemical recurrence following radical prostatectomy and we validated this model in two external data sets. METHODS We analyzed prostate specimens from patients undergoing radical prostatectomy at Hartford Hospital between 2008 and 2011. RNA isolated from formalin-fixed paraffin-embedded prostates was hybridized to a custom Affymetrix microarray. Regularized (least absolute shrinkage and selection operator [Lasso]) Cox regression was performed with cross-validation to identify a model that incorporated gene expression and clinical factors to predict biochemical recurrence, defined as postoperative prostate-specific antigen (PSA) > 0.2 ng/ml or receipt of triggered salvage treatment. Model performance was assessed using time-dependent receiver operating curve (ROC) curves and survival plots. RESULTS A total of 606 prostate specimens with gene expression and both pre- and postoperative PSA data were available for analysis. We identified a model that included Gleason grade and stage as well as five genes (CNRIP1, endoplasmic reticulum protein 44 [ERP44], metaxin-2 [MTX2], Ras homolog family member U [RHOU], and OXR1). Using the Lasso method, we determined that the five gene model independently predicted biochemical recurrence better than a model that included Gleason grade and tumor stage alone. The time-dependent ROCAUC for the five gene signature including Gleason grade and tumor stage was 0.868 compared to an AUC of 0.767 when Gleason grade and tumor stage were included alone. Low and high-risk groups displayed significant differences in their recurrence-free survival curves. The predictive model was subsequently validated on two independent data sets identified through the Gene Expression Omnibus. The model included genes (RHOU, MTX2, and ERP44) that have previously been implicated in prostate cancer biology. CONCLUSIONS Expression of a small number of genes is associated with an increased risk of biochemical recurrence independent of classical pathological hallmarks.
Collapse
Affiliation(s)
- John P Corradi
- Hartford Hospital Research Program, Hartford Hospital, Hartford, Connecticut, USA
| | | | - Ilene Staff
- Hartford Hospital Research Program, Hartford Hospital, Hartford, Connecticut, USA
| | - Joseph Tortora
- Hartford Hospital Research Program, Hartford Hospital, Hartford, Connecticut, USA
| | - Andrew Salner
- Hartford Healthcare Cancer Institute, Hartford, Connecticut, USA
| | - Tara McLaughlin
- Hartford Hospital Research Program, Hartford Hospital, Hartford, Connecticut, USA
| | - Joseph Wagner
- Urology Division, Hartford Healthcare Medical Group, Hartford Hospital, Hartford, Connecticut, USA
| |
Collapse
|
34
|
Tilki D, Chen MH, Wu J, Huland H, Graefen M, Wiegel T, Böhmer D, Mohamad O, Cowan JE, Feng FY, Carroll PR, Trock BJ, Partin AW, D'Amico AV. Adjuvant Versus Early Salvage Radiation Therapy for Men at High Risk for Recurrence Following Radical Prostatectomy for Prostate Cancer and the Risk of Death. J Clin Oncol 2021; 39:2284-2293. [PMID: 34086480 DOI: 10.1200/jco.20.03714] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Adjuvant compared with early salvage radiation therapy (sRT) following radical prostatectomy (RP) has not been shown to reduce progression-free survival in randomized controlled trials. However, these trials might have missed a benefit in men with adverse pathology at RP given that these men were under-represented and immortal time bias might have been present; herein, we investigate this possibility. METHODS We evaluated the impact of adjuvant versus early sRT on all-cause mortality (ACM) risk in men with adverse pathology defined as positive pelvic lymph nodes (pN1) or pGleason score 8-10 prostate cancer (PC) and disease extending beyond the prostate (pT3/4). We used a treatment propensity score to minimize potential treatment selection bias when estimating the causal effect of adjuvant versus early sRT on ACM risk and a sensitivity analysis to assess the impact that varying definitions of adverse pathology had on ACM risk adjusting for age at RP, PC prognostic factors, site, and the time-dependent use of post-RP androgen deprivation therapy. RESULTS After a median follow-up (interquartile range) of 8.16 (6.00-12.10) years, of the 26,118 men in the study cohort, 2,104 (8.06%) died, of which 539 (25.62%) were from PC. After excluding men with a persistent prostate-specific antigen, adjuvant compared with early sRT was associated with a significantly lower ACM risk among men with adverse pathology at RP when men with pN1 PC were excluded (0.33 [0.13-0.85]; P = .02) or included (0.66 [0.44-0.99]; P = .04). CONCLUSION Adjuvant radiation therapy should be considered in men with pN1 or pGleason score 8 to 10 and pT3/4 PC given the possibility that a significant reduction in ACM risk exists.
Collapse
Affiliation(s)
- Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital-Hamburg-Eppendorf, Hamburg, Germany.,Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ming-Hui Chen
- Department of Statistics, University of Connecticut, Storrs, CT
| | - Jing Wu
- Department of Computer Science and Statistics, University of Rhode Island, Kingston, RI
| | - Hartwig Huland
- Martini-Klinik Prostate Cancer Center, University Hospital-Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Graefen
- Martini-Klinik Prostate Cancer Center, University Hospital-Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Wiegel
- Department of Radio Oncology, University Hospital Ulm, Ulm, Germany
| | - Dirk Böhmer
- Department of Radiation Oncology, Charité University Hospital, Berlin, Germany
| | - Osama Mohamad
- Department of Radiation Oncology, University of San Francisco, San Francisco, CA
| | - Janet E Cowan
- Department of Urology, University of San Francisco, San Francisco, CA
| | - Felix Y Feng
- Department of Radiation Oncology, University of San Francisco, San Francisco, CA.,Department of Urology, University of San Francisco, San Francisco, CA
| | - Peter R Carroll
- Department of Urology, University of San Francisco, San Francisco, CA
| | - Bruce J Trock
- Division of Epidemiology, Brady Urological Institute at Johns Hopkins Medical Institution, Baltimore, MD
| | - Alan W Partin
- Department of Urology, Brady Urological Institute at Johns Hopkins Medical Institution, Baltimore, MD
| | - Anthony V D'Amico
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, MA
| |
Collapse
|
35
|
Campbell SR, Tom MC, Agrawal S, Efstathiou JA, Michalski JM, Abramowitz MC, Pollack A, Spratt DE, Hearn JWD, Stephans KL, Gao T, Li J, Tendulkar RD. Integrating Prostate-specific Antigen Kinetics into Contemporary Predictive Nomograms of Salvage Radiotherapy After Radical Prostatectomy. Eur Urol Oncol 2021; 5:304-313. [PMID: 34016556 DOI: 10.1016/j.euo.2021.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Salvage radiotherapy (SRT) is an established treatment for men with biochemical recurrence following radical prostatectomy (RP). There are several risk factors associated with adverse outcomes; however, the value of postoperative prostate-specific antigen (PSA) kinetics is less clear in the ultrasensitive PSA era. OBJECTIVE To characterize the impact of PSA kinetics on outcomes following SRT and generate nomograms to aid in identifying patients with an increased risk of adverse clinical outcomes. DESIGN, SETTING, AND PARTICIPANTS A multi-institutional analysis was conducted of 1005 patients with prostate cancer treated with SRT after RP, with a median follow-up of 5 years. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Variables examined include immediate postoperative PSA, postoperative PSA doubling time (DT), and pre-SRT PSA, in addition to previously identified predictive factors. Multivariable survival analyses were completed using Fine-Gray competing risk regression. Rates of biochemical failure (BF), distant metastasis (DM), and prostate cancer-specific mortality (PCSM) were estimated by the cumulative incidence method. Nomograms were generated from multivariable competing risk regression with bootstrap cross-validation. RESULTS AND LIMITATIONS Factors associated with BF after SRT include PSA DT <6 mo, initial postoperative PSA ≥0.2 ng/ml, higher pre-SRT PSA, lack of androgen deprivation therapy, a higher Gleason score (GS), negative margins, seminal vesicle invasion, lack of pelvic nodal radiation, radiation total dose <66 Gy, a longer RP to SRT interval, and older age (p < 0.05 for each). Factors associated with DM include PSA DT <6 mo, pre-SRT PSA, a higher GS, and negative margins. Factors associated with PCSM include PSA DT not calculable or <6 mo and a higher GS. Nomograms were generated to estimate the risks of BF (concordance index [CI] 0.74), DM (CI 0.77), and PCSM (CI 0.77). Limitations include retrospective nature, broad treatment eras, institutional variations, and multiple methods available for the estimation of PSA DT. CONCLUSIONS Postoperative PSA kinetics, particularly pre-SRT PSA and PSA DT, are strongly associated with adverse oncologic outcomes following SRT and should be considered in management decisions. PATIENT SUMMARY In this report of men with prostate cancer who developed a prostate-specific antigen (PSA) recurrence after prostatectomy, we found that PSA levels after surgery and how quickly a PSA level doubles significantly impact the chance of prostate cancer recurrence after salvage radiation therapy. Based on this information, we created a tool to calculate a man's chance of cancer recurrence after salvage radiation therapy, and these estimations can be used to discuss whether additional treatment with radiation should be considered.
Collapse
Affiliation(s)
- Shauna R Campbell
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Martin C Tom
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Shree Agrawal
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jason A Efstathiou
- Department of Radiation Oncology, Dana-Farber/Harvard Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Jeff M Michalski
- Department of Radiation Oncology, Siteman Cancer Center, Washington University, St. Louis, MO, USA
| | - Matthew C Abramowitz
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Alan Pollack
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals, Cleveland, Ohio
| | - Jason W D Hearn
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kevin L Stephans
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Tianming Gao
- Global Medical Affairs Statistics, AbbVie, Chicago, IL, USA
| | - Jianbo Li
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Rahul D Tendulkar
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
36
|
Zhang L, Li Y, Wang X, Ping Y, Wang D, Cao Y, Dai Y, Liu W, Tao Z. Five-gene signature associating with Gleason score serve as novel biomarkers for identifying early recurring events and contributing to early diagnosis for Prostate Adenocarcinoma. J Cancer 2021; 12:3626-3647. [PMID: 33995639 PMCID: PMC8120165 DOI: 10.7150/jca.52170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Compared to non-recurrent type, recurrent prostate adenocarcinoma (PCa) is highly fatal, and significantly shortens the survival time of affected patients. Early and accurate laboratory diagnosis is particularly important in identifying patients at high risk of recurrence, necessary for additional systemic intervention. We aimed to develop efficient and accurate diagnostic and prognostic biomarkers for new PCa following radical therapy. Methods: We identified differentially expressed genes (DEGs) and clinicopathological data of PCa patients from Gene Expression Omnibus (GEO) datasets and The Cancer Genome Atlas (TCGA) repositories. We then uncovered the most relevant clinical traits and genes modules associated with PCa prognosis using the Weighted gene correlation network analysis (WGCNA). Univariate Cox regression analysis and multivariate Cox proportional hazards (Cox-PH) models were performed to identify candidate gene signatures related to Disease-Free Interval (DFI). Data for internal and external cohorts were utilized to test and validate the accuracy and clinical utility of the prognostic models. Results: We constructed and validated an accurate and reliable model for predicting the prognosis of PCa using 5 Gleason score-associated gene signatures (ZNF695, CENPA, TROAP, BIRC5 and KIF20A). The ROC and Kaplan-Meier analysis revealed the model was highly accurate in diagnosing and predicting the recurrence and metastases of PCa. The accuracy of the model was validated using the calibration curves based on internal TCGA cohort and external GEO cohort. Using the model, patients could be prognostically stratified in to various groups including TNM classification and Gleason score. Multivariate analysis revealed the model could independently predict the prognosis of PCa patients and its utility was superior to that of clinicopathological characteristics. In addition, we fund the expression of the 5 gene signatures strongly and positively correlated with tumor purity but negatively correlated with infiltration CD8+ T cells to the tumor microenvironment. Conclusions: A 5 gene signatures can accurately be used in the diagnosis and prediction of PCa prognosis. Thus this can guide the treatment and management prostate adenocarcinoma.
Collapse
Affiliation(s)
- Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yu Li
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui 233030, China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Ping
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Danhua Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
37
|
Genomic Strategies to Personalize Use of Androgen Deprivation Therapy With Radiotherapy. ACTA ACUST UNITED AC 2021; 26:13-20. [PMID: 31977380 DOI: 10.1097/ppo.0000000000000419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The use of combination RT and androgen deprivation therapy in many prostate cancer curative-intent treatment scenarios is supported by level 1 evidence. However, in our current clinical paradigm, we have no ability to determine a priori which patients truly benefit from combination therapy and therefore apply the combination RT and androgen deprivation therapy intensification strategy to all patients, which results in overtreatment or undertreatment of the majority of our patients. Genomics has the ability to more deeply and objectively characterize the disease, in turn refining our prognostication capabilities and enabling the individualization of treatments. We review the commercially available prostate cancer genomic tests, focusing on those able to predict patient outcomes following radiotherapy or guide radiotherapy treatment decisions.
Collapse
|
38
|
Affiliation(s)
- Sean E McGuire
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
39
|
Ferro M, Lucarelli G, de Cobelli O, Del Giudice F, Musi G, Mistretta FA, Luzzago S, Busetto GM, Buonerba C, Sciarra A, Conti S, Porreca A, Battaglia M, Ditonno P, Manfredi M, Fiori C, Porpiglia F, Terracciano D. The emerging landscape of tumor marker panels for the identification of aggressive prostate cancer: the perspective through bibliometric analysis of an Italian translational working group in uro-oncology. Minerva Urol Nephrol 2021; 73:442-451. [PMID: 33769016 DOI: 10.23736/s2724-6051.21.04098-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Molecular heterogeneity and availability of different therapeutic strategies are relevant clinical features of prostate cancer. On this basis, there is an urgent need to identify prognostic and predictive biomarkers for an individualized therapeutic approach. In this context, researchers focused their attention on biomarkers able to discriminate potential life-threatening from organ-confined disease. Such biomarker could provide aid in clinical decision making, helping to choose the treatment which ensures the best results in terms of patient survival and quality of life. To address this need, many new laboratory tests have been proposed, with a clear tendency to use panels of combined biomarkers. In this review we evaluate current data on the application in clinical practice of the most promising laboratory tests: Phi, 4K score and Stockholm 3 as circulating biomarkers, Mi-prostate score, Exo DX Prostate and Select MD-X as urinary biomarkers, Confirm MDx, Oncotype Dx, Prolaris and Decipher as tissue biomarkers. In particular, the ability of these tests in the identification of clinically significant PCa and their potential use for precision medicine have been explored in this review.
Collapse
Affiliation(s)
- Matteo Ferro
- Department of Urology, IEO European Institute of Oncology, Milan, Italy
| | - Giuseppe Lucarelli
- Unit of Urology, Andrology and Kidney Transplantation, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Ottavio de Cobelli
- Department of Urology, IEO European Institute of Oncology, Milan, Italy.,Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
| | | | - Gennaro Musi
- Department of Urology, IEO European Institute of Oncology, Milan, Italy
| | | | - Stefano Luzzago
- Department of Urology, IEO European Institute of Oncology, Milan, Italy
| | | | - Carlo Buonerba
- Service of Medical Oncology, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Simon Conti
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Angelo Porreca
- Department of Urology, Abano Terme Hospital, Padua, Italy
| | - Michele Battaglia
- Unit of Urology, Andrology and Kidney Transplantation, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Pasquale Ditonno
- Unit of Urology, Andrology and Kidney Transplantation, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Matteo Manfredi
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Cristian Fiori
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Francesco Porpiglia
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, Federico II University, Naples, Italy -
| |
Collapse
|
40
|
White C, Staff I, McLaughlin T, Tortora J, Pinto K, Gangakhedkar A, Champagne A, Wagner J. Does post prostatectomy decipher score predict biochemical recurrence and impact care? World J Urol 2021; 39:3281-3286. [PMID: 33743058 DOI: 10.1007/s00345-021-03661-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/06/2021] [Indexed: 11/26/2022] Open
Abstract
PURPOSE To examine the ability of the Decipher test to predict early biochemical recurrence after radical prostatectomy and to impact clinical decisions in advance of metastasis and death. METHODS We identified Decipher tests ordered after radical prostatectomy for adverse pathology in men treated for prostate cancer between 1/1/14 and 8/31/18. Biochemical recurrence was defined as prostate-specific antigen > 0.02 ng/mL. Decipher score is reported as lower risk (< 0.6) and higher risk ≥ 0.60). Kaplan-Meier analysis was used to examine the relationship between Decipher score and time to biochemical recurrence (months). Cox regression was used to analyze the relationship between Decipher score and time to biochemical recurrence while controlling for a number of clinical characteristics. Secondary analyses focused on a subset of men with prostate-specific antigen > 0.02 and < 0.20 ng/mL to determine if high-risk Decipher scores were associated with receipt of salvage treatment. RESULTS A total of 203 cases were analyzed: 37.9% and 62.1% had lower and higher risk Decipher scores respectively, and 56.2% had a biochemical recurrence. Median (inter-quartile range) follow-up was 20 (13.5, 25.3) months. Decipher score was significantly associated with time to biochemical recurrence (p = 0.027) while in the secondary analyses, high-risk Decipher scores (≥ 0.60) were associated with salvage treatment (p = 0.018). Stage category and Decipher score were significant predictors of time from elevated PSA to salvage treatment in the secondary analyses. CONCLUSION While it might not contribute statistically, Decipher score can be clinically useful in helping patients reach treatment decisions.
Collapse
Affiliation(s)
- Christine White
- Urology Division, Hartford Healthcare Medical Group, Hartford Hospital, 85 Seymour Street, Suite 416, Hartford, CT, 06106, USA
| | - Ilene Staff
- Hartford Hospital Research Program, Hartford Hospital, Hartford, CT, 06106, USA
| | - Tara McLaughlin
- Urology Division, Hartford Healthcare Medical Group, Hartford Hospital, 85 Seymour Street, Suite 416, Hartford, CT, 06106, USA.
| | - Joseph Tortora
- Hartford Hospital Research Program, Hartford Hospital, Hartford, CT, 06106, USA
| | - Kevin Pinto
- Urology Division, Hartford Healthcare Medical Group, Hartford Hospital, 85 Seymour Street, Suite 416, Hartford, CT, 06106, USA
| | - Akshay Gangakhedkar
- Urology Division, Hartford Healthcare Medical Group, Hartford Hospital, 85 Seymour Street, Suite 416, Hartford, CT, 06106, USA
| | - Alison Champagne
- Hartford Hospital Research Program, Hartford Hospital, Hartford, CT, 06106, USA
| | - Joseph Wagner
- Urology Division, Hartford Healthcare Medical Group, Hartford Hospital, 85 Seymour Street, Suite 416, Hartford, CT, 06106, USA
| |
Collapse
|
41
|
Morlacco A, Modonutti D, Motterle G, Martino F, Dal Moro F, Novara G. Nomograms in Urologic Oncology: Lights and Shadows. J Clin Med 2021; 10:jcm10050980. [PMID: 33801184 PMCID: PMC7957873 DOI: 10.3390/jcm10050980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/08/2021] [Accepted: 02/20/2021] [Indexed: 12/29/2022] Open
Abstract
Decision-making in urologic oncology involves integrating multiple clinical data to provide an answer to the needs of a single patient. Although the practice of medicine has always been an “art” involving experience, clinical data, scientific evidence and judgment, the creation of specialties and subspecialties has multiplied the challenges faced every day by physicians. In the last decades, with the field of urologic oncology becoming more and more complex, there has been a rise in tools capable of compounding several pieces of information and supporting clinical judgment and experience when approaching a difficult decision. The vast majority of these tools provide a risk of a certain event based on various information integrated in a mathematical model. Specifically, most decision-making tools in the field of urologic focus on the preoperative or postoperative phase and provide a prognostic or predictive risk assessment based on the available clinical and pathological data. More recently, imaging and genomic features started to be incorporated in these models in order to improve their accuracy. Genomic classifiers, look-up tables, regression trees, risk-stratification tools and nomograms are all examples of this effort. Nomograms are by far the most frequently used in clinical practice, but are also among the most controversial of these tools. This critical, narrative review will focus on the use, diffusion and limitations of nomograms in the field of urologic oncology.
Collapse
Affiliation(s)
- Alessandro Morlacco
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Daniele Modonutti
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Giovanni Motterle
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Francesca Martino
- Department of Nephrology, Dialysis and Kidney Transplant, International Renal Research Institute, San Bortolo Hospital, 36100 Vicenza, Italy;
| | - Fabrizio Dal Moro
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
| | - Giacomo Novara
- Urology Unit, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, 35128 Padua, Italy; (A.M.); (D.M.); (G.M.); (F.D.M.)
- Correspondence: or ; Tel.: +39-049-821-1250; Fax: +39-049-821-8757
| |
Collapse
|
42
|
Shahait M, Liu VYT, Vapiwala N, Lal P, Kim J, Trabulsi EJ, Huang H, Davicioni E, Thompson DJS, Spratt D, Den RB, Lee DI. Impact of Decipher on use of post‐operative radiotherapy: Individual patient analysis of two prospective registries. BJUI COMPASS 2021; 2:267-274. [PMID: 35475294 PMCID: PMC8988525 DOI: 10.1002/bco2.70] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 11/10/2022] Open
Abstract
Objective To assess the association between Genomic Classifier (GC)‐risk group and post‐radical prostatectomy treatment in clinical practice. Methods Two prospective observational cohorts of men with prostate cancer (PCa) who underwent RP in two referral centers and had GC testing post‐prostatectomy between 2013 and 2018 were included. The primary endpoint of the study was to assess the association between GC‐risk group and time to secondary therapy. Univariable (UVA) and multivariable (MVA) Cox proportional hazards models were constructed to assess the association between GC‐risk group and time to receipt of secondary therapy after RP, where secondary therapy is defined as receiving either RT or ADT after RP. Results A total of 398 patients are included in the analysis. Patients with high‐GC risk were more likely to receive any secondary therapy (OR: 6.84) compared to patients with low/intermediate‐GC risk. The proportion of high‐GC risk patients receiving RT at 2 years post‐RP was 31.5%, compared to only 6.3% among the low/intermediate‐GC risk patients. Conclusion This study demonstrates that physicians in routine practice used GC to identify high risk patients who might benefit the most from secondary treatment. As such, GC score was independent predictor of receipt of secondary treatment.
Collapse
Affiliation(s)
| | | | - Neha Vapiwala
- Department of Radiation Oncology Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Priti Lal
- Department of Pathology Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Jessica Kim
- Department of Surgery University of Pennsylvania Philadelphia PA USA
| | | | | | | | | | | | - Robert B. Den
- Thomas Jefferson University Hospital Philadelphia PA USA
| | - David I. Lee
- Department of Surgery University of Pennsylvania Philadelphia PA USA
- Penn Urology Penn Presbyterian Medical Center Philadelphia PA USA
| |
Collapse
|
43
|
Moris L, Devos G, Van den Broeck T, Milonas D, Albersen M, Berghen C, De Meerleer G, Devlies W, Everaerts W, Gevaert T, Van Poppel H, Claessens F, Joniau S. Current and emerging therapies for localized high-risk prostate cancer. Expert Rev Anticancer Ther 2020; 21:267-282. [PMID: 33225759 DOI: 10.1080/14737140.2021.1852932] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Despite progress in the field of high-risk localized prostate cancer (HRPCa) treatments, high-risk patients treated with curative intent are at increased risk of biochemical recurrence, metastatic progression and cancer-related death. The optimal treatment strategy remains a topic of debate. This review provides an overview of the current and investigational therapeutic options for HRPCa.Areas covered: A PubMed search was performed for papers on the current perspectives on the multimodality treatment of HRPCa. We focus on both primary local treatment as well as systemic treatment options. Finally, relevant ongoing trials focusing on systemic treatments (including [neo]adjuvant treatments) enrolling at least 50 patients were retrieved, to highlight ongoing research and treatment optimization.Expert opinion: Disease progression in HRPCa patients is driven by local tumor extension and subclinical metastases. Therefore, the main treatment concept is a multimodal approach targeting the primary tumor with extended surgery or RT with long-term ADT and simultaneously targeting micro-metastatic deposits. However, there is still room for optimization. Upcoming clinical trials comparing surgery versus RT as local treatment, trials with (neo)adjuvant chemotherapy or androgen receptor signaling inhibitors will likely change the treatment landscape. However, a multimodal treatment strategy will stay as the cornerstone in the treatment of HRPCa.
Collapse
Affiliation(s)
- Lisa Moris
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Gaëtan Devos
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | | | - Daimantas Milonas
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Department of Urology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Charlien Berghen
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Gert De Meerleer
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Thomas Gevaert
- Department of Pathology, Catholic University Leuven, Belgium
| | - Hendrik Van Poppel
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| |
Collapse
|
44
|
Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers. I. Molecular Biomarkers in Prostate Cancer. Am J Surg Pathol 2020; 44:e15-e29. [PMID: 32044806 DOI: 10.1097/pas.0000000000001450] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The combined clinical and molecular heterogeneity of prostate cancer necessitates the use of prognostic, predictive, and diagnostic biomarkers to assist the clinician with treatment selection. The pathologist plays a critical role in guiding molecular biomarker testing in prostate cancer and requires a thorough knowledge of the current testing options. In the setting of clinically localized prostate cancer, prognostic biomarkers such as Ki-67 labeling, PTEN loss or mRNA-based genomic signatures can be useful to help determine whether definitive therapy is required. In the setting of advanced disease, predictive biomarkers, such as the presence of DNA repair deficiency mediated by BRCA2 loss or mismatch repair gene defects, may suggest the utility of poly-ADP ribosylase inhibition or immune checkpoint blockade. Finally, androgen receptor-related biomarkers or diagnostic biomarkers indicating the presence of small cell neuroendocrine prostate cancer may help guide the use of androgen receptor signaling inhibitors and chemotherapy. In this review, we examine the current evidence for several prognostic, predictive and diagnostic tissue-based molecular biomarkers in prostate cancer management. For each assay, we summarize a recent survey of the International Society of Urology Pathology (ISUP) members on current testing practices and include recommendations for testing that emerged from the ISUP Working Group on Molecular Pathology of Prostate Cancer and the 2019 Consultation Conference on Molecular Pathology of Urogenital Cancers.
Collapse
|
45
|
Shelan M, Aebersold D, Ghadjar P. [Early salvage radiation therapy of the prostate bed appears to be equally effective compared to adjuvant radiation therapy after radical prostatectomy]. Strahlenther Onkol 2020; 196:406-409. [PMID: 32060583 DOI: 10.1007/s00066-020-01591-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- M Shelan
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Schweiz
| | - D Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Schweiz
| | - P Ghadjar
- Klinik für Radioonkologie und Strahlentherapie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany, Augustenburger Platz 1, 13353, Berlin, Deutschland.
| |
Collapse
|
46
|
Delgadillo R, Ford JC, Abramowitz MC, Dal Pra A, Pollack A, Stoyanova R. The role of radiomics in prostate cancer radiotherapy. Strahlenther Onkol 2020; 196:900-912. [PMID: 32821953 PMCID: PMC7545508 DOI: 10.1007/s00066-020-01679-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/07/2020] [Indexed: 12/24/2022]
Abstract
"Radiomics," as it refers to the extraction and analysis of a large number of advanced quantitative radiological features from medical images using high-throughput methods, is perfectly suited as an engine for effectively sifting through the multiple series of prostate images from before, during, and after radiotherapy (RT). Multiparametric (mp)MRI, planning CT, and cone beam CT (CBCT) routinely acquired throughout RT and the radiomics pipeline are developed for extraction of thousands of variables. Radiomics data are in a format that is appropriate for building descriptive and predictive models relating image features to diagnostic, prognostic, or predictive information. Prediction of Gleason score, the histopathologic cancer grade, has been the mainstay of the radiomic efforts in prostate cancer. While Gleason score (GS) is still the best predictor of treatment outcome, there are other novel applications of quantitative imaging that are tailored to RT. In this review, we summarize the radiomics efforts and discuss several promising concepts such as delta-radiomics and radiogenomics for utilizing image features for assessment of the aggressiveness of prostate cancer and its outcome. We also discuss opportunities for quantitative imaging with the advance of instrumentation in MRI-guided therapies.
Collapse
Affiliation(s)
- Rodrigo Delgadillo
- Department of Radiation Oncology, University of Miami Miller School of Medicine, 1121 NW 14th St, 33136, Miami, FL, USA
| | - John C Ford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, 1121 NW 14th St, 33136, Miami, FL, USA
| | - Matthew C Abramowitz
- Department of Radiation Oncology, University of Miami Miller School of Medicine, 1121 NW 14th St, 33136, Miami, FL, USA
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami Miller School of Medicine, 1121 NW 14th St, 33136, Miami, FL, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, 1121 NW 14th St, 33136, Miami, FL, USA
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, 1121 NW 14th St, 33136, Miami, FL, USA.
| |
Collapse
|
47
|
Darrell CM, Montironi R, Paner GP. Potential biomarkers and risk assessment models to enhance the tumor-node-metastasis (TNM) staging classification of urologic cancers. Expert Rev Mol Diagn 2020; 20:921-932. [PMID: 32876523 DOI: 10.1080/14737159.2020.1816827] [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: 10/23/2022]
Abstract
INTRODUCTION The anatomic-based TNM classification is considered the benchmark in cancer staging and has been regularly updated since its inception. In the current era of precision medicine, the added intention for future TNM modifications is to heighten its impact in the more 'personalized' level of cancer care. In urologic cancers, this goal may be achieved by incorporating 'non-anatomic' factors into TNM, such as biomarkers (e.g. gene alterations, molecular subtypes, genomic classifiers) and risk assessment models (e.g. nomogram, look-up table), while maintaining the anatomic extent as the foundation of staging. These different prognosticators can be combined and integrated, may serve as substratifiers for T, N, or M categories, and perhaps, incorporated as elements in TNM stage groupings to enhance their prognostic capability in urologic cancers. AREAS COVERED This review highlights candidate biomarkers and risk assessment models that can be explored to potentially improve TNM prognostication of bladder, prostate, kidney, and testicular cancers. EXPERT OPINION Recent advances in molecular analysis have increased the understanding of the genomic, transcriptomic, and epigenetic features for biomarker use in prognostication of urologic cancers, which together with the available risk assessment models, may complement and overcome the limitations of the traditional TNM staging.
Collapse
Affiliation(s)
- Caitlin M Darrell
- Departments of Pathology, Section of Urology, University of Chicago , Chicago, IL, USA
| | - Rodolfo Montironi
- School of Medicine, Section of Pathological Anatomy, Polytechnic University of the Marche Region , Ancona, Italy
| | - Gladell P Paner
- Departments of Pathology, Section of Urology, University of Chicago , Chicago, IL, USA.,Departments of Surgery, Section of Urology, University of Chicago , Chicago, IL, USA
| |
Collapse
|
48
|
Saoud R, Heidar NA, Cimadamore A, Paner GP. Incorporating Prognostic Biomarkers into Risk Assessment Models and TNM Staging for Prostate Cancer. Cells 2020; 9:E2116. [PMID: 32957584 PMCID: PMC7564222 DOI: 10.3390/cells9092116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/06/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
In current practice, prostate cancer staging alone is not sufficient to adequately assess the patient's prognosis and plan the management strategies. Multiple clinicopathological parameters and risk tools for prostate cancer have been developed over the past decades to better characterize the disease and provide an enhanced assessment of prognosis. Herein, we review novel prognostic biomarkers and their integration into risk assessment models for prostate cancer focusing on their capability to help avoid unnecessary imaging studies, biopsies and diagnosis of low risk prostate cancers, to help in the decision-making process between active surveillance and treatment intervention, and to predict recurrence after radical prostatectomy. There is an imperative need of reliable biomarkers to stratify prostate cancer patients that may benefit from different management approaches. The integration of biomarkers panel with risk assessment models appears to improve prostate cancer diagnosis and management. However, integration of novel genomic biomarkers in future prognostic models requires further validation in their clinical efficacy, standardization, and cost-effectiveness in routine application.
Collapse
Affiliation(s)
- Ragheed Saoud
- Department of Surgery (Section of Urology), University of Chicago, Chicago, IL 60637, USA;
| | - Nassib Abou Heidar
- Department of Surgery (Division of Urology), American University of Beirut Medical Center, Beirut 11-0236, Lebanon;
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, 60126 Ancona, Italy;
| | - Gladell P. Paner
- Department of Surgery (Section of Urology), University of Chicago, Chicago, IL 60637, USA;
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
49
|
Mohler JL, Antonarakis ES, Armstrong AJ, D'Amico AV, Davis BJ, Dorff T, Eastham JA, Enke CA, Farrington TA, Higano CS, Horwitz EM, Hurwitz M, Ippolito JE, Kane CJ, Kuettel MR, Lang JM, McKenney J, Netto G, Penson DF, Plimack ER, Pow-Sang JM, Pugh TJ, Richey S, Roach M, Rosenfeld S, Schaeffer E, Shabsigh A, Small EJ, Spratt DE, Srinivas S, Tward J, Shead DA, Freedman-Cass DA. Prostate Cancer, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 17:479-505. [PMID: 31085757 DOI: 10.6004/jnccn.2019.0023] [Citation(s) in RCA: 882] [Impact Index Per Article: 176.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The NCCN Guidelines for Prostate Cancer include recommendations regarding diagnosis, risk stratification and workup, treatment options for localized disease, and management of recurrent and advanced disease for clinicians who treat patients with prostate cancer. The portions of the guidelines included herein focus on the roles of germline and somatic genetic testing, risk stratification with nomograms and tumor multigene molecular testing, androgen deprivation therapy, secondary hormonal therapy, chemotherapy, and immunotherapy in patients with prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Joseph E Ippolito
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Jesse McKenney
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - George Netto
- University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | | | | | - Sylvia Richey
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Mack Roach
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | - Edward Schaeffer
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Ahmad Shabsigh
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Eric J Small
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | | | - Jonathan Tward
- Huntsman Cancer Institute at the University of Utah; and
| | | | | |
Collapse
|
50
|
Cullen J, Kuo HC, Shan J, Lu R, Aboushwareb T, Van Den Eeden SK. The 17-Gene Genomic Prostate Score Test as a Predictor of Outcomes in Men with Unfavorable Intermediate Risk Prostate Cancer. Urology 2020; 143:103-111. [PMID: 32525077 DOI: 10.1016/j.urology.2020.05.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/12/2020] [Accepted: 05/16/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To evaluate the association of the Genomic Prostate Score (GPS) assay result with biochemical recurrence (BCR), distant metastases (DM), and prostate-specific death (PCD) in unfavorable intermediate (UFI) risk prostate cancer patients. The GPS assay is used to help guide management decisions for newly diagnosed low and favorable intermediate (FI) risk disease. METHODS GPS results from 2 studies (Center for Prostate Disease Research [CPDR]; Kaiser Permanente Northern California [KPNC]) in men treated with radical prostatectomy were analyzed to determine associations of the GPS result with BCR, DM, and PCD in UFI risk disease. Analyses included 299 intermediate risk prostate patients, 175 of whom had UFI risk disease (KPNC = 103; CPDR = 72). RESULTS The GPS result as a dichotomous value (≤40 vs >40) was a significant predictor of BCR in UFI patients in multivariate analyses (hazard ratio [HR] 6.0; 95% confidence interval [CI] 2.0-22.4; P = .0035; CPDR). The GPS result was a strong predictor of all 3 endpoints in multivariate analyses (BCR HR 7.1; 95% CI 5.7-8.8; P < .0001; DM HR 5.4; 95% CI 3.8-7.8; P < .0001; PCD HR 3.4; 95% CI 1.5-8.9; P = .006; KPNC). UFI patients with GPS >40 had outcomes consistent with high-risk disease, whereas UFI patients with GPS ≤40 had outcomes similar to FI risk patients (CPDR/KPNC). CONCLUSIONS The GPS result was a strong independent predictor of BCR, DM, and PCD in intermediate risk prostate cancer. UFI patients with GPS >40 have a poor prognosis and may benefit from additional therapeutic options.
Collapse
Affiliation(s)
- Jennifer Cullen
- Cancer Population Sciences, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH.
| | - Huai-Ching Kuo
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, MD; Henry M. Jackson Foundation, Bethesda, MD
| | - Jun Shan
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Ruixiao Lu
- Genomic Health Inc., an Exact Sciences Corporation, Redwood City, CA
| | - Tamer Aboushwareb
- Genomic Health Inc., an Exact Sciences Corporation, Redwood City, CA
| | | |
Collapse
|