1
|
Patel KR, Nguyen PL, Proudfoot JA, Liu Y, Pra AD, Spratt DE, Pollack A, Sandler HM, Efstathiou JA, Lawton C, Simko JP, Rosenthal SA, Zeitzer KL, Mendez LC, Hartford AC, Hall WA, Desai AB, Pugh SL, Davicioni E, Tran PT, Feng FY. Biopsy-based Basal-luminal Subtyping Classifier in High-risk Prostate Cancer: A Combined Analysis of the NRG Oncology/RTOG 9202, 9413, and 9902 Phase 3 Trials. Eur Urol Oncol 2024:S2588-9311(24)00246-3. [PMID: 39542826 DOI: 10.1016/j.euo.2024.10.017] [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: 09/06/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND AND OBJECTIVE Long-term (LT) androgen deprivation therapy (ADT) has been found to be beneficial to patients with high-risk prostate cancer (PCa). However, administration of LT-ADT to all patients with high-risk PCa may lead to overtreatment. Enhanced risk stratification using genomic classifiers (such as the recently developed prostate subtyping classifier [PSC]) might be useful. This study aims to characterize the prognostic and predictive ability of the PSC in patients with high-risk PCa undergoing radiotherapy long-term (LT; 24-28 mo) versus short-term (ST; 4 mo) ADT. METHODS Biopsy samples from three randomized, phase 3 trials-NRG/RTOG 9202, 9413, and 9902-were classified as either PSC basal or luminal. The prognostic and predictive values of PSC for each oncologic endpoint (biochemical failure [BF], distant metastasis [DM], metastasis-free survival [MFS], PCa-specific mortality [PCSM], overall survival [OS]) and other cause-mortality (OCM) were assessed with Cox proportional hazards (MFS, OCM, and OS), Fine-Gray (BF, DM, and PCSM), and restricted mean survival time (RMST) models. KEY FINDINGS AND LIMITATIONS On a multivariable analysis, the basal subtype was found to have a worse prognosis for MFS (hazard ratio [HR] 1.8 [1.3-2.5], p < 0.001), PCSM (subdistribution HR 2.4 [95% confidence interval {CI} 1.4-4.1], p = 0.001), and OS (HR 1.8 [1.3-2.6], p < 0.001). Ten-year PCSM was 15% better for the luminal subtype than for the basal subtype (11% [95% CI 6-15%] vs 26% [95% CI 17-35%]). A significant interaction between ADT duration (LT vs ST) and PSC subtype (basal vs luminal) was observed for PCSM (pinteraction = 0.008), leading to the observation that 10-yr PCSM was improved with LT-ADT only in patients with basal-type tumors (5% [95% CI 0-11%] vs 42% [29-56%], p < 0.001). Improvements in 10-yr RMST with LT-ADT were greater for basal tumors for oncologic endpoints with the exception of OCM. CONCLUSIONS AND CLINICAL IMPLICATIONS PSC is both a prognostic and a predictive biomarker for patients who benefit from LT-ADT. PSC subtypes may be used to personalize ADT recommendations for patients with high-risk PCa, pending further validation in a prospective study. PATIENT SUMMARY In this study, we tried to understand the usefulness of a new genomic test in patients with high-risk, nonmetastatic prostate cancer who underwent radiation therapy and hormonal therapy (HT). We found that this test can help determine a patient's prognosis (eg, a patient's chance of having the cancer return) and, more importantly, personalize treatment decisions by understanding which patients may benefit from long-term HT. This has the potential to save many patients who may not benefit from prolonged HT from "overtreatment" or the unnecessary side effects of such treatment.
Collapse
Affiliation(s)
- Krishnan R Patel
- Radiation Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | | | - Yang Liu
- Veracyte, Inc, South San Francisco, CA, USA
| | - Alan Dal Pra
- University of Miami Cancer Center, Miami, FL, USA
| | | | - Alan Pollack
- University of Miami Cancer Center, Miami, FL, USA
| | | | | | | | | | - Seth A Rosenthal
- Sutter Cancer Centers Radiation Oncology Services, Roseville, CA, USA
| | - Kenneth L Zeitzer
- Einstein Medical Center, Accruals for Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | | | - Alan C Hartford
- Dartmouth-Hitchcock Medical Center/Norris Cotton Cancer Center, Lebanon, NH, USA
| | - William A Hall
- Froedtert and the Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Stephanie L Pugh
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA, USA; ACR, American College of Radiology, Philadelphia, PA, USA
| | | | - Phuoc T Tran
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD, USA.
| | - Felix Y Feng
- UCSF Medical Center-Mission Bay, San Francisco, CA, USA.
| |
Collapse
|
2
|
Parker CC, Clarke NW, Cook AD, Petersen PM, Catton CN, Cross WR, Kynaston H, Persad RA, Saad F, Logue J, Payne H, Amos C, Bower L, Raman R, Sayers I, Worlding J, Parulekar WR, Parmar MKB, Sydes MR. Randomised Trial of No, Short-term, or Long-term Androgen Deprivation Therapy with Postoperative Radiotherapy After Radical Prostatectomy: Results from the Three-way Comparison of RADICALS-HD (NCT00541047). Eur Urol 2024; 86:422-430. [PMID: 39217077 DOI: 10.1016/j.eururo.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND OBJECTIVE The use and duration of androgen deprivation therapy (ADT) with postoperative radiotherapy (RT) have been uncertain. RADICALS-HD compared adding no ("None"), 6-months ("Short"), or 24-mo ("Long") ADT to study efficacy in the long term. METHODS Participants with prostate cancer were indicated for postoperative RT and agreed randomisation between all durations. ADT was allocated for 0, 6, or 24 mo. The primary outcome measure (OM) was metastasis-free survival (MFS). The secondary OMs included freedom from distant metastasis, overall survival, and initiation of nonprotocol ADT. Sample size was determined by two-way comparisons. Analyses followed standard time-to-event approaches and intention-to-treat principles. KEY FINDINGS AND LIMITATIONS Between 2007 and 2015, 492 participants were randomised one of three groups: 166 None, 164 Short, and 162 Long. The median age at randomisation was 66 yr; Gleason scores at surgery were as follows: <7 = 64 (13%), 3+4 = 229 (47%), 4+3 = 127 (26%), and 8+ = 72 (15%); T3b was 112 (23%); and T4 was 5 (1%). The median follow-up was 9.0 yr and, with MFS events reported for 89 participants (32 None, 31 Short, and 26 Long), there was no evidence of difference in MFS overall (logrank p = 0.98), and, for Long versus None, hazard ratio = 0.948 (95% confidence interval 0.54-1.68). After 10 yr, 80% None, 77% Short, and 81% Long patients were alive without metastatic disease. The three-way randomisation was not powered to conventional levels for assessment, yet provides a fair comparison. CONCLUSIONS AND CLINICAL IMPLICATIONS Long-term outcomes after radical prostatectomy are usually favourable. In those indicated for postoperative RT and considered suitable for no, short-term, or long-term ADT, there was no evidence of improvement with addition of ADT. Future research should focus on patients at a higher risk of metastases in whom improvements are required more urgently.
Collapse
Affiliation(s)
- Chris C Parker
- Royal Marsden NHS Foundation Trust, Sutton, UK; The Institute of Cancer Research, Sutton, UK
| | - Noel W Clarke
- Department of Urology, The Christie and Salford Royal Hospitals, Manchester, UK; The University of Manchester, Manchester, UK
| | - Adrian D Cook
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Peter M Petersen
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Charles N Catton
- Department of Radiation Oncology, Princess Margaret, Cancer Centre, University Health Network, Toronto, ON, Canada
| | - William R Cross
- Department of Urology, St James's University Hospital, Leeds, UK
| | - Howard Kynaston
- Division of Cancer & Genetics, Cardiff University Medical School, Cardiff, UK
| | - Raj A Persad
- Department of Urology, Bristol Urological Institute, Bristol, UK
| | - Fred Saad
- Department of Urology, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | | | | | - Claire Amos
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Lorna Bower
- The Institute of Cancer Research, Sutton, UK; Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Rakesh Raman
- Kent Oncology Centre, Kent & Canterbury Hospital, Canterbury, UK
| | - Ian Sayers
- Deanesly Centre, New Cross Hospital, Wolverhampton, UK
| | - Jane Worlding
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Wendy R Parulekar
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Mahesh K B Parmar
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK.
| |
Collapse
|
3
|
González-San Segundo C, López Campos F, Gómez Iturriaga A, Rodríguez A, Olivera J, Duque-Santana V, Sancho G, Henríquez I, Conde AJ, Valero J, Maldonado X, Glaria L, Caballero B, Sanmamed N, Mases J, Boladeras-Inglada AM, Montijano M, Santos M, Álvarez A, Martínez JI, Couñago F. A randomised trial of short- vs long-term androgen deprivation with salvage radiotherapy for biochemical failure following radical prostatectomy: URONCOR 06-24. BJU Int 2024; 134:568-577. [PMID: 39041411 DOI: 10.1111/bju.16484] [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] [Indexed: 07/24/2024]
Abstract
BACKGROUND Salvage radiotherapy (SRT) and androgen-deprivation therapy (ADT) are widely used in routine clinical practice to treat patients with prostate cancer who develop biochemical recurrence (BCR) after radical prostatectomy (RP). However, there is no standard-of-care consensus on optimal duration ADT. Investigators propose three distinct risk groups in patients with prostate cancer treated with SRT in order to better define the indications and duration of ADT combined with SRT. STUDY DESIGN The URONCOR 06-24 trial (ClinicalTrials.gov identifier NCT05781217) is a prospective, multicentre, randomised, open-label, phase III, clinical trial. The aim of the trial is to determine the impact of short-term (6 months) vs long-term (24 months) ADT in combination with SRT on distant metastasis-free survival (MFS) in patients with prostate cancer with BCR after RP (intermediate and high risk). ENDPOINTS The primary endpoint is 5-year MFS rates in patients with prostate cancer treated with long- vs short-term ADT in combination with SRT. Secondary objectives are biochemical-relapse free interval, pelvic progression-free survival, time to start of systemic treatment, time to castration resistance, cancer-specific survival, overall survival, acute and late toxicity, and quality of life. METHODS AND ANALYSIS Total of 534 patients will be randomised 1:1 to ADT 6 months or ADT 24 months with a luteinizing hormone-releasing hormone analogue in combination with SRT, stratified by risk group and pathological lymph node status. ETHICS AND DISSEMINATION The study is conducted under the guiding principles of the World Medical Association Declaration of Helsinki. The results will be disseminated at research conferences and in peer-reviewed journals. TRIAL REGISTRATION NUMBER EudraCT number 2021-006975-41.
Collapse
Affiliation(s)
| | - Fernando López Campos
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Department of Radiation Oncology, Genesis Care Hospital Vithas La Milagrosa, Madrid, Spain
| | - Alfonso Gómez Iturriaga
- Department of Radiation Oncology, Hospital Universitario Cruces, Biobizkaia Health Research Institute Basque Country University UPV/EHU, Barakaldo, Spain
| | - Aurora Rodríguez
- Department of Radiation Oncology, Hospital Ruber Internacional, Madrid, Spain
| | - Jesús Olivera
- Department of Radiation Oncology, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | | | - Gemma Sancho
- Department of Radiation Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Iván Henríquez
- Department of Radiation Oncology, Hospital Universitario Sant Joan, Reus, Tarragona, Spain
| | - Antonio José Conde
- Department of Radiation Oncology, Hospital Universitario La Fe, Valencia, Spain
| | - Jeannette Valero
- Department of Radiation Oncology, Hospital Universitario HM San Chinarro, Madrid, Spain
| | - Xavier Maldonado
- Department of Radiation Oncology, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Luis Glaria
- Department of Radiation Oncology, Hospital Universitario La Paz, Madrid, Spain
| | - Begoña Caballero
- Department of Radiation Oncology, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Noelia Sanmamed
- Department of Radiation Oncology, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Joel Mases
- Department of Radiation Oncology, Hospital Universitario Clínic de Barcelona, Barcelona, Spain
| | | | - Miguel Montijano
- Department of Urology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Marina Santos
- Department of Radiation Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Ana Álvarez
- Department of Radiation Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Juan I Martínez
- Department of Urology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Felipe Couñago
- Department of Radiation Oncology, GenesisCare Hospital San Francisco de Asís, Hospital Vithas La Milagrosa, Madrid, Spain
- Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|
4
|
Pommier P, Xie W, Ravi P, Carrie C, Dignam JJ, Feng F, Sargos P, Sommer SG, Spratt DE, Tombal B, Van Poppel H, Sweeney C. Prognostic factors in post-prostatectomy salvage radiotherapy setting with and without hormonotherapy: An individual patient data analysis of randomized trials from ICECaP database. Radiother Oncol 2024:110532. [PMID: 39278317 DOI: 10.1016/j.radonc.2024.110532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Early salvage radiotherapy (SRT) is the standard of care for biochemical recurrence post-prostatectomy but outcomes are heterogeneous. OBJECTIVE To develop a risk scoring system based on relevant standard-of-care clinico-pathological prognostic factors for patients treated with SRT with and without hormonal therapy (HT). DESIGN, SETTING, AND PARTICIPANTS The Intermediate Clinical Endpoints in Cancer of the Prostate (ICECaP) database included three randomized trials (Individual patients' data from 1647 subjects) assessing SRT (GETUG-AFU-16; NRG/RTOG-9601, and a subset of EORTC-22911). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Outcomes were clinical progression (CP). metastasis free-survival (MFS) and overall survival (OS). Clinico-pathological factors, including pathological Gleason Score (GS), PSA at SRT start, margin status, persistent PSA post-RP and time from RP to SRT were evaluated by multivariable models stratified by type of treatment. RESULTS AND LIMITATIONS On multivariable analysis PSA ≥ 0.5 ng/mL at SRT start, GS ≥ 8 and negative margin status were the three strongest prognostic factors. Three prognostic groups defined by number of these risk features (high risk: 2 or 3; intermediate risk: 1 and low risk: 0) were strongly associated with OS, MFS and CP outcomes with SRT alone or with HT. This prognostic group definition was also relevant for patients with persistent PSA post RP and for patients treated < 1 year from RP to SRT and with and without HT. CONCLUSION A risk score for patients receiving SRT with or without HT, using three standard-of-care clinico-pathological risk factors provides refined prognostic information for individual patient counselling. PATIENT SUMMARY By using a composite score of pathology grading (Gleason Score), PSA at start of salvage radiation and margin status data, physicians can provide patients with more refined information on the risk of a second relapse after receiving radiation to the prostate bed after a prostatectomy for a rising or persistent PSA, both with and without hormonal therapy.
Collapse
Affiliation(s)
- Pascal Pommier
- Department of Radiation Oncology, Curie Institute, Paris, France.
| | - Wanling Xie
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Praful Ravi
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Christian Carrie
- Department of Radiation Oncology, Centre Léon Bérard, Lyon, France
| | | | - Felix Feng
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonié, Bordeaux, France
| | - Silke Gillessen Sommer
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Christopher Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
5
|
Heutlinger O, Azizi A, Harada G, Harris JP, Daneshvar M, Gin G, Uchio E, Mar N, Rezazadeh A, Seyedin SN. Socioeconomic Barriers to Receiving Early Salvage Radiotherapy for Locally Advanced Prostate Adenocarcinoma: A Retrospective Single-Center Study. Cureus 2024; 16:e68945. [PMID: 39381448 PMCID: PMC11460723 DOI: 10.7759/cureus.68945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 10/10/2024] Open
Abstract
Purpose This study aimed to identify factors associated with delays in initiating early salvage radiation therapy in prostate cancer patients with prostate-specific antigen (PSA) failure after prostatectomy. Methods We conducted a single-institution, retrospective study of patients receiving salvage radiation therapy after radical prostatectomy from 2011 to 2022. Patient demographics and clinical data were examined to identify factors that may have influenced the time to start of radiation therapy after surgery. Utilizing a PSA cut off of 0.25 ng/ml or less, we classified patients as receiving either early "PSA low" or late "PSA high" salvage therapy depending on their PSA at the time of initiating treatment. Results Of the 81 patients evaluated, the median age was 61.9 years (IQR 57.9 - 66.5), with most presenting with pT3 (65.4%), Grade Group 2 disease (35.8%), and positive margins 55%). Median PSA at salvage radiation therapy commencement was 0.30 ng/mL (0.18 - 0.48). 40 patients completed early salvage and 41 patients completed late salvage in the overall cohort. A significant association was found between patient insurance carrier and pre-radiation PSA levels. Patients with HMO (Health Maintenance Organization) or PPO (Preferred Provider Organization) insurance were more likely to complete late salvage radiation compared to non-managed Medicare patients (HMO OR 4.0, p <0.05 & PPO OR 3.3 p <0.05 vs non-managed Medicare). All uninsured patients in the cohort received late salvage radiation. Conclusions Insurance type was significantly associated with the timing of salvage radiation therapy post-prostatectomy, suggesting a relationship with providers requiring prior authorization (HMO and PPO coverage). This study supports proper PSA surveillance, in particular for those with HMO or PPO coverage.
Collapse
Affiliation(s)
- Olivia Heutlinger
- Radiation Oncology, University of California Irvine School of Medicine, Irvine, USA
| | - Armon Azizi
- Radiation Oncology, University of California Irvine School of Medicine, Irvine, USA
| | - Garrett Harada
- Radiation Oncology, University of California Irvine Medical Center, Orange, USA
| | - Jeremy P Harris
- Radiation Oncology, University of California Irvine Medical Center, Orange, USA
| | - Michael Daneshvar
- Urology, University of California Irvine Medical Center, Orange, USA
| | - Greg Gin
- Urology, University of California Irvine Medical Center, Orange, USA
| | - Edward Uchio
- Urology, University of California Irvine Medical Center, Orange, USA
| | - Nataliya Mar
- Hematology and Oncology, University of California Irvine Medical Center, Orange, USA
| | - Arash Rezazadeh
- Hematology and Medical Oncology, University of California Irvine Medical Center, Orange, USA
| | - Steven N Seyedin
- Radiation Oncology, University of California San Francisco Medical Center, San Francisco, USA
| |
Collapse
|
6
|
Shelan M, Achard V, Appiagyei F, Mose L, Zilli T, Fankhauser CD, Zamboglou C, Mohamad O, Aebersold DM, Cathomas R. Role of enzalutamide in primary and recurrent non-metastatic hormone sensitive prostate cancer: a systematic review of prospective clinical trials. Prostate Cancer Prostatic Dis 2024; 27:422-431. [PMID: 38589645 PMCID: PMC11319196 DOI: 10.1038/s41391-024-00829-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Enzalutamide, a second-generation androgen receptor inhibitor, is indicated for the treatment of metastatic disease, as well as in the treatment of non-metastatic castration resistant prostate cancer (PCa). This systematic review aims to determine outcomes and toxicity in patients with non-metastatic castration sensitive prostate cancer (nmCSPC) treated with enzalutamide in the primary or salvage settings. METHOD We performed a systematic review focusing on the role of Enzalutamide in the treatment of nmCSPC, using the PubMed/Medline database. Articles focusing on androgen receptor inhibitors in nmCSPC were included, while articles discussing exclusively metastatic or castration-resistant PCa were excluded. RESULTS The initial search retrieved 401 articles, of which 15 underwent a thorough assessment for relevance. Ultimately, 12 studies with pertinent outcomes were meticulously examined. Among these, seven studies were dedicated to the investigation of enzalutamide in the primary setting, while the remaining five publications specifically addressed its use in salvage settings. Regardless of the treatment setting, our data revealed two distinct therapeutic strategies. The first advocates for the substitution of enzalutamide for androgen deprivation therapy (ADT), based on the premise of achieving equivalent, if not superior, oncological outcomes while minimizing treatment-related toxicity. The second, adopting a more conventional approach, entails augmenting the effectiveness of ADT by incorporating enzalutamide. CONCLUSION Enzalutamide has considerable potential as a therapeutic strategy for nmCSPC, either used alone or in combination with ADT in the primary or in the salvage settings. The use of enzalutamide instead of ADT is an appealing strategy. However, more trials will be required to further understand the efficacy and side-effect profile of enzalutamide monotherapy.
Collapse
Affiliation(s)
- Mohamed Shelan
- Department of Radiation Oncology, Inselspital Bern, University of Bern, Bern, Switzerland.
| | - Vérane Achard
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Radiation Oncology, HFR Fribourg, Villars-sur-Glâne, Switzerland
| | - Felix Appiagyei
- Department of Radiation Oncology, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Lucas Mose
- Department of Radiation Oncology, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Thomas Zilli
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Radiation Oncology, Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Christian D Fankhauser
- Department of Urology, Luzerner Kantonsspital, University of Lucerne, Lucerne, Switzerland
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Oncology Center, University Hospital of the European University, Limassol, Cyprus
| | - Osama Mohamad
- Department of Genito-urinary Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Richard Cathomas
- Department of Oncology/Hematology, Kantonsspital Graubünden, Chur, Switzerland
| |
Collapse
|
7
|
Weiner AB, Kakani P, Armstrong AJ, Bossi A, Cornford P, Feng F, Kanabur P, Karnes RJ, Mckay RR, Morgan TM, Schaeffer EM, Shore N, Tree AC, Spratt DE. Risk Stratification of Patients with Recurrence After Primary Treatment for Prostate Cancer: A Systematic Review. Eur Urol 2024; 86:200-210. [PMID: 38782697 DOI: 10.1016/j.eururo.2024.04.034] [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: 02/12/2024] [Revised: 04/04/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND OBJECTIVE Biochemical recurrence (BCR) after primary definitive treatment for prostate cancer (PCa) is a heterogeneous disease state. While BCR is associated with worse oncologic outcomes, risk factors that impact outcomes can vary significantly, necessitating avenues for risk stratification. We sought to identify prognostic risk factors at the time of recurrence after primary radical prostatectomy or radiotherapy, and prior to salvage treatment(s), associated with adverse oncologic outcomes. METHODS We performed a systematic review of prospective studies in EMBASE, MEDLINE, and ClinicalTrials.gov (from January 1, 2000 to October 16, 2023) according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines (CRD42023466330). We reviewed the factors associated with oncologic outcomes among patients with BCR after primary definitive treatment. KEY FINDINGS AND LIMITATIONS A total of 37 studies were included (total n = 10 632), 25 after prostatectomy (total n = 9010) and 12 after radiotherapy (total n = 1622). Following recurrence after prostatectomy, factors associated with adverse outcomes include higher pathologic T stage and grade group, negative surgical margins, shorter prostate-specific antigen doubling time (PSADT), higher prostate-specific antigen (PSA) prior to salvage treatment, shorter time to recurrence, the 22-gene tumor RNA signature, and recurrence location on molecular imaging. After recurrence following radiotherapy, factors associated with adverse outcomes include a shorter time to recurrence, and shorter PSADT or higher PSA velocity. Grade group, T stage, and prior short-term hormone therapy (4-6 mo) were not clearly associated with adverse outcomes, although sample size and follow-up were generally limited compared with postprostatectomy data. CONCLUSIONS AND CLINICAL IMPLICATIONS This work highlights the recommendations and level of evidence for risk stratifying patients with PCa recurrence, and can be used as a benchmark for personalizing salvage treatment based on prognostics.
Collapse
Affiliation(s)
- Adam B Weiner
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Preeti Kakani
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Andrew J Armstrong
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute Center for Prostate and Urologic Cancer, Duke University, Durham, NC, USA
| | - Alberto Bossi
- Amethyst Radiotherapy Group, La Garenne Colombes, France
| | | | - Felix Feng
- Department of Radiation Oncology, University of California at San Francisco, San Francisco, CA, USA
| | - Pratik Kanabur
- Department of Urology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Rana R Mckay
- Department of Medicine, Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Todd M Morgan
- Department of Urology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Edward M Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Alison C Tree
- Department of Radiotherapy, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
8
|
Vera G, Rojas PA, Black JB, San Francisco IF. Usefulness of Tissue Biomarkers versus Prostate-Specific Membrane Antigen-Positron Emission Tomography for Prostate Cancer Biochemical Recurrence after Radical Prostatectomy. Cancers (Basel) 2024; 16:2879. [PMID: 39199648 PMCID: PMC11352583 DOI: 10.3390/cancers16162879] [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: 07/17/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Despite curative-intent local therapy, approximately 27% to 53% of prostate cancer (PCa) patients experience prostate-specific antigen (PSA) recurrence, known as biochemical recurrence (BCR). BCR significantly raises the risk of PCa-related morbidity and mortality, yet there is no consensus on optimal management. Prostate-specific membrane antigen-positron emission tomography (PSMA PET) has emerged as highly sensitive imaging, distinguishing local recurrences from distant metastases, crucially influencing treatment decisions. Genomic biomarkers such as Decipher, Prolaris, and Oncotype DX contribute to refining recurrence risk profiles, guiding decisions on intensifying adjuvant therapies, like radiotherapy and androgen deprivation therapy (ADT). This review assesses PSMA PET and biomarker utility in post-radical prostatectomy BCR scenarios, highlighting their impact on clinical decision-making. Despite their promising roles, the routine integration of biomarkers is limited by availability and cost, requiring further evidence. PSMA PET remains indispensable for restaging and treatment evaluation in these patients. Integrating biomarkers and PSMA PET promises to optimize personalized management strategies for BCR, though more comprehensive consensus-building studies are needed to define their standardized utility in clinical practice.
Collapse
Affiliation(s)
- Gabriela Vera
- Servicio de Urología, Complejo Asistencial Dr. Sotero del Rio, Santiago 8207257, Chile; (G.V.); (P.A.R.)
| | - Pablo A. Rojas
- Servicio de Urología, Complejo Asistencial Dr. Sotero del Rio, Santiago 8207257, Chile; (G.V.); (P.A.R.)
| | - Joseph B. Black
- Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215-5400, USA;
| | - Ignacio F. San Francisco
- Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215-5400, USA;
| |
Collapse
|
9
|
Mattes MD. Overview of Radiation Therapy in the Management of Localized and Metastatic Prostate Cancer. Curr Urol Rep 2024; 25:181-192. [PMID: 38861238 DOI: 10.1007/s11934-024-01217-5] [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] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
PURPOSE OF REVIEW The goal is to describe the evolution of radiation therapy (RT) utilization in the management of localized and metastatic prostate cancer. RECENT FINDINGS Long term data for a variety of hypofractionated definitive RT dose-fractionation schemes has matured, allowing patients and providers many standard-of-care options to choose from. Post-prostatectomy, adjuvant RT has largely been replaced by an early salvage approach. Multiparametric MRI and PSMA PET have enabled increasingly targeted RT delivery to the prostate and oligometastatic tumors. Areas of active investigation include determining the value of proton beam therapy and perirectal spacers, and optimally incorporate genomic tumor profiling and next generation hormonal therapies with RT in the curative setting. The use of radiation therapy to treat prostate cancer is rapidly evolving. In the coming years, there will be continued improvements in a variety of areas to enhance the value of RT in multidisciplinary prostate cancer management.
Collapse
Affiliation(s)
- Malcolm D Mattes
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
10
|
Armstrong WR, Kishan AU, Booker KM, Grogan TR, Elashoff D, Lam EC, Clark KJ, Steinberg ML, Fendler WP, Hope TA, Nickols NG, Czernin J, Calais J. Impact of Prostate-specific Membrane Antigen Positron Emission Tomography/Computed Tomography on Prostate Cancer Salvage Radiotherapy Management: Results from a Prospective Multicenter Randomized Phase 3 Trial (PSMA-SRT NCT03582774). Eur Urol 2024; 86:52-60. [PMID: 38290964 DOI: 10.1016/j.eururo.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/12/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND AND OBJECTIVE Both imaging and several prognostic factors inform the planning of salvage radiotherapy (SRT). Prostate-specific membrane antigen positron emission tomography (PSMA-PET) can localize disease unseen by other imaging modalities. The main objective of the study was to evaluate the impact of PSMA-PET on biochemical recurrence-free survival rate after SRT. METHODS This prospective randomized, controlled, phase 3 clinical trial randomized 193 patients with biochemical recurrence of prostate cancer after radical prostatectomy to proceed with SRT (control arm, n = 90) or undergo a PSMA-PET/computed tomography (CT) scan prior to SRT planning (investigational arm, n = 103) from June 2018 to August 2020. Any other approved imaging modalities were allowed in both arms (including fluciclovine-PET). This is a secondary endpoint analysis: impact of PSMA-PET on SRT planning. Case-report forms were sent to referring radiation oncologists to collect the management plans before randomization and after completion of SRT. The relative frequency (%) of management changes within each arm were compared using chi-square and Fisher's exact tests. KEY FINDINGS AND LIMITATIONS The delivered SRT plan was available in 178/193 patients (92.2%;76/90 control [84.4%] and 102/103 PSMA-PET [99%]). Median prostate-specific antigen levels at enrollment was 0.30 ng/ml (interquartile range [IQR] 0.19-0.91) in the control arm and 0.23 ng/ml (IQR 0.15-0.54) in the PSMA-PET arm. Fluciclovine-PET was used in 33/76 (43%) in the control arm. PSMA-PET localized recurrence(s) in 38/102 (37%): nine of 102 (9%) outside of the pelvis (M1), 16/102 (16%) in the pelvic LNs (N1, with or without local recurrence), and 13/102 (13%) in the prostate fossa only. There was a 23% difference (95% confidence interval [CI] 9-35%, p = 0.002) of frequency of major changes between the control arm (22% [17/76]) and the PSMA-PET intervention arm (45%[46/102]). Of the major changes in the intervention group, 33/46 (72%) were deemed related to PSMA-PET. There was a 17.6% difference (95% CI 5.4-28.5%, p = 0.005) of treatment escalation frequency between the control arm (nine of 76 [12%]) and the intervention arm (30/102 [29%]). Treatment de-escalation occurred in the control and intervention arms in eight of 76 (10.5%) and 12/102 (11.8%) patients, and mixed changes in zero of 76 (0%) and four of 102 (3.9%) patients, respectively. CONCLUSIONS AND CLINICAL IMPLICATIONS In this prospective randomized phase 3 study, PSMA-PET findings provided information that initiated major management changes to SRT planning in 33/102 (33%) patients. The final readout of the primary endpoint planned in 2025 may provide evidence on whether these changes result in improved outcomes. PATIENT SUMMARY Prostate-specific membrane antigen positron emission tomography leads to management changes in one-third of patients receiving salvage radiotherapy for post-radical prostatectomy biochemical recurrence of prostate cancer.
Collapse
Affiliation(s)
- Wesley R Armstrong
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Kiara M Booker
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Tristan R Grogan
- Department of Medicine Statistics Core (DOMStat), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David Elashoff
- Department of Medicine Statistics Core (DOMStat), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ethan C Lam
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kevyn J Clark
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael L Steinberg
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Wolfgang P Fendler
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK) - University Hospital Essen, Essen, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Nicholas G Nickols
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA; Department of Radiation Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Shore ND, Moul JW, Pienta KJ, Czernin J, King MT, Freedland SJ. Biochemical recurrence in patients with prostate cancer after primary definitive therapy: treatment based on risk stratification. Prostate Cancer Prostatic Dis 2024; 27:192-201. [PMID: 37679602 PMCID: PMC11096125 DOI: 10.1038/s41391-023-00712-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Nearly one-third of patients with prostate cancer (PCa) experience biochemical recurrence (BCR) after primary definitive treatment. BCR increases the risk of distant metastasis and mortality in patients with prognostically unfavorable features. These patients are best managed with a tailored treatment strategy incorporating risk stratification using clinicopathological factors, next-generation imaging, and genomic testing. OBJECTIVE This narrative review examines the utility of risk stratification for the management of patients with BCR in the context of clinical trial data, referencing the latest recommendations by European and US medical societies. METHODS PubMed was searched for relevant studies published through May 21 2023 on treatment of patients with BCR after radical prostatectomy (RP) or external beam radiotherapy (EBRT). RESULTS European and US guidelines support the risk-stratified management of BCR. Post-RP, salvage EBRT (with or without androgen deprivation therapy [ADT]) is an accepted treatment option for patients with BCR. Post-EBRT, local salvage therapies (RP, cryotherapy, high-intensity focused ultrasound, stereotactic body radiotherapy, and low-dose-rate and high-dose-rate brachytherapy) have demonstrated comparable relapse-free survival rates but differing adverse event profiles, short and long term. Local salvage therapies should be used for local-only relapses while ADT should be considered for regional or distant relapses. In practice, patients often receive ADT, with varying guidance for intermittent ADT vs. continuous ADT, due to consideration of quality-of-life effects. CONCLUSIONS Despite a lack of consensus for BCR treatment among guideline associations and medical societies, risk stratification of patients is essential for personalized treatment approaches, as it allows for an informed selection of therapeutic strategies and estimation of adverse events. In lower-risk disease, observation is recommended while in higher-risk disease, after failed repeat local therapy, ADT and/or clinical trial enrollment may be appropriate. Results from ongoing clinical studies of patients with BCR should provide consensus for management.
Collapse
Affiliation(s)
- Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Judd W Moul
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | | | - Johannes Czernin
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Martin T King
- Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J Freedland
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Veterans Affairs Medical Center, Durham, NC, USA.
| |
Collapse
|
12
|
Parker CC, Kynaston H, Cook AD, Clarke NW, Catton CN, Cross WR, Petersen PM, Persad RA, Pugh CA, Saad F, Logue J, Payne H, Bower LC, Brawley C, Rauchenberger M, Barkati M, Bottomley DM, Brasso K, Chung HT, Chung PWM, Conroy R, Falconer A, Ford V, Goh CL, Heath CM, James ND, Kim-Sing C, Kodavatiganti R, Malone SC, Morris SL, Nabid A, Ong AD, Raman R, Rodda S, Wells P, Worlding J, Parulekar WR, Parmar MKB, Sydes MR. Duration of androgen deprivation therapy with postoperative radiotherapy for prostate cancer: a comparison of long-course versus short-course androgen deprivation therapy in the RADICALS-HD randomised trial. Lancet 2024; 403:2416-2425. [PMID: 38763153 PMCID: PMC7616389 DOI: 10.1016/s0140-6736(24)00549-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Previous evidence supports androgen deprivation therapy (ADT) with primary radiotherapy as initial treatment for intermediate-risk and high-risk localised prostate cancer. However, the use and optimal duration of ADT with postoperative radiotherapy after radical prostatectomy remains uncertain. METHODS RADICALS-HD was a randomised controlled trial of ADT duration within the RADICALS protocol. Here, we report on the comparison of short-course versus long-course ADT. Key eligibility criteria were indication for radiotherapy after previous radical prostatectomy for prostate cancer, prostate-specific antigen less than 5 ng/mL, absence of metastatic disease, and written consent. Participants were randomly assigned (1:1) to add 6 months of ADT (short-course ADT) or 24 months of ADT (long-course ADT) to radiotherapy, using subcutaneous gonadotrophin-releasing hormone analogue (monthly in the short-course ADT group and 3-monthly in the long-course ADT group), daily oral bicalutamide monotherapy 150 mg, or monthly subcutaneous degarelix. Randomisation was done centrally through minimisation with a random element, stratified by Gleason score, positive margins, radiotherapy timing, planned radiotherapy schedule, and planned type of ADT, in a computerised system. The allocated treatment was not masked. The primary outcome measure was metastasis-free survival, defined as metastasis arising from prostate cancer or death from any cause. The comparison had more than 80% power with two-sided α of 5% to detect an absolute increase in 10-year metastasis-free survival from 75% to 81% (hazard ratio [HR] 0·72). Standard time-to-event analyses were used. Analyses followed intention-to-treat principle. The trial is registered with the ISRCTN registry, ISRCTN40814031, and ClinicalTrials.gov, NCT00541047. FINDINGS Between Jan 30, 2008, and July 7, 2015, 1523 patients (median age 65 years, IQR 60-69) were randomly assigned to receive short-course ADT (n=761) or long-course ADT (n=762) in addition to postoperative radiotherapy at 138 centres in Canada, Denmark, Ireland, and the UK. With a median follow-up of 8·9 years (7·0-10·0), 313 metastasis-free survival events were reported overall (174 in the short-course ADT group and 139 in the long-course ADT group; HR 0·773 [95% CI 0·612-0·975]; p=0·029). 10-year metastasis-free survival was 71·9% (95% CI 67·6-75·7) in the short-course ADT group and 78·1% (74·2-81·5) in the long-course ADT group. Toxicity of grade 3 or higher was reported for 105 (14%) of 753 participants in the short-course ADT group and 142 (19%) of 757 participants in the long-course ADT group (p=0·025), with no treatment-related deaths. INTERPRETATION Compared with adding 6 months of ADT, adding 24 months of ADT improved metastasis-free survival in people receiving postoperative radiotherapy. For individuals who can accept the additional duration of adverse effects, long-course ADT should be offered with postoperative radiotherapy. FUNDING Cancer Research UK, UK Research and Innovation (formerly Medical Research Council), and Canadian Cancer Society.
Collapse
Affiliation(s)
- Chris C Parker
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - Howard Kynaston
- Division of Cancer and Genetics, Cardiff University Medical School, Cardiff, UK
| | - Adrian D Cook
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Noel W Clarke
- Department of Urology, The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK; Department of Urology, Salford Royal Hospital, Salford, UK
| | | | - William R Cross
- Department of Urology, St James's University Hospital, Leeds, UK
| | - Peter M Petersen
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Cheryl A Pugh
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Fred Saad
- Department of Urology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - John Logue
- Department of Urology, The Christie NHS Foundation Trust, Manchester, UK
| | | | - Lorna C Bower
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK; Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Chris Brawley
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Mary Rauchenberger
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Maroie Barkati
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - David M Bottomley
- Department of Clinical Oncology, St James's University Hospital, Leeds, UK
| | - Klaus Brasso
- Department of Urology, Copenhagen Prostate Cancer Center, Rigshospitalet, Copenhagen, Denmark
| | - Hans T Chung
- Department of Radiation Oncology, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Peter W M Chung
- Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Ruth Conroy
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | | | - Vicky Ford
- Royal Devon and Exeter University NHS Foundation Trust, Exeter, UK
| | - Chee L Goh
- Royal Surrey County Hospital, Guildford, UK
| | - Catherine M Heath
- Department of Clinical Oncology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Nicholas D James
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - Charmaine Kim-Sing
- Department of Radiation Oncology, BC Cancer-Vancouver, Vancouver, BC, Canada
| | - Ravi Kodavatiganti
- Glan Clwyd Hospital, Betsi Cadwaladr University Health Board, Bangor, UK
| | - Shawn C Malone
- The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | | | - Abdenour Nabid
- Service de Radio-Oncologie, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Aldrich D Ong
- Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Rakesh Raman
- Kent Oncology Centre, Kent and Canterbury Hospital, Canterbury, UK
| | - Sree Rodda
- Bradford Teaching Hospitals, Bradford, UK
| | - Paula Wells
- Barts Cancer Centre, St Bartholomew's Hospital, London, UK
| | - Jane Worlding
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Wendy R Parulekar
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Mahesh K B Parmar
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK.
| |
Collapse
|
13
|
Pollack A, Pra AD. Androgen deprivation therapy combined with postoperative radiotherapy for prostate cancer management. Lancet 2024; 403:2353-2355. [PMID: 38763152 DOI: 10.1016/s0140-6736(24)00802-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 05/21/2024]
Affiliation(s)
- Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA.
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| |
Collapse
|
14
|
Deng Y, Zhang C, Yu H, Chen G, Peng X, Li Y, Feng Z, Shi W, Bai X, Gou X, Liu N. AAT resistance-related AC007405.2 and AL354989.1 as novel diagnostic and prognostic markers in prostate cancer. Aging (Albany NY) 2024; 16:7249-7266. [PMID: 38643469 PMCID: PMC11087092 DOI: 10.18632/aging.205754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/28/2024] [Indexed: 04/22/2024]
Abstract
OBJECTIVE Prostate cancer (PCa) is the second disease threatening men's health, and anti-androgen therapy (AAT) is a primary approach for treating this condition. Increasing evidence suggests that long non-coding RNAs (lncRNAs) play crucial roles in the development of PCa and the process of AAT resistance. The objective of this study is to utilize bioinformatics methods to excavate lncRNAs association with AAT resistance and investigate their biological functions. METHODS AAT resistance-related risk score model (ARR-RSM) was established by multivariate Cox analysis. Paired clinical tissue samples of 36 PCa patients and 42 blood samples from patients with PSA over 4 ng/ml were collected to verify the ARR-RSM. In vitro, RT-qPCR, CCK-8 and clone formation assays were displayed to verify the expression and function of AL354989.1 and AC007405.2. RESULTS Pearson correlation analysis identified 996 lncRNAs were associated with AAT resistance (ARR-LncRs). ARR-RSM was established using multivariate Cox regression analysis, and PCa patients were divided into high-risk and low-risk groups. High-risk patients showed increased expression of AL354989.1 and AC007405.2 had poorer prognoses. The high-risk score correlated with advanced T-stage and N-stage. The AUC of ARR-RSM outperformed tPSA in diagnosing PCa. Silencing of AC007405.2 and AL354989.1 inhibited PCa cells proliferation and AAT resistance. CONCLUSIONS In this study, we have discovered the clinical significance of AC007405.2 and AL354989.1 in predicting the prognosis and diagnosing PCa patients. Furthermore, we have confirmed their correlation with various clinical features. These findings provide potential targets for PCa treatment and a novel diagnostic and predictive indicator for precise PCa diagnosis.
Collapse
Affiliation(s)
- Yuanzhong Deng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
| | - Chunlin Zhang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Haitao Yu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Guo Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Xiang Peng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Yang Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Zhenwei Feng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Wei Shi
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Xuesong Bai
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Yuzhong, Chongqing, China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
| | - Nian Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, China
| |
Collapse
|
15
|
von Eyben FE, Kairemo K, Kapp DS. Prostate-Specific Antigen as an Ultrasensitive Biomarker for Patients with Early Recurrent Prostate Cancer: How Low Shall We Go? A Systematic Review. Biomedicines 2024; 12:822. [PMID: 38672176 PMCID: PMC11048591 DOI: 10.3390/biomedicines12040822] [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: 09/15/2023] [Revised: 12/25/2023] [Accepted: 03/05/2024] [Indexed: 04/28/2024] Open
Abstract
Serum prostate-specific antigen (PSA) needs to be monitored with ultrasensitive PSA assays (uPSAs) for oncologists to be able to start salvage radiotherapy (SRT) while PSA is <0.5 µg/L for patients with prostate cancer (PCa) relapsing after a radical prostatectomy (RP). Our systematic review (SR) aimed to summarize uPSAs for patients with localized PCa. The SR was registered as InPLASY2023110084. We searched for studies on Google Scholar, PUBMED and reference lists of reviews and studies. We only included studies on uPSAs published in English and excluded studies of women, animals, sarcoidosis and reviews. Of the 115 included studies, 39 reported PSA assay methods and 76 reported clinical findings. Of 67,479 patients, 14,965 developed PSA recurrence (PSAR) and 2663 died. Extremely low PSA nadir and early developments of PSA separated PSAR-prone from non-PSAR-prone patients (cumulative p value 3.7 × 1012). RP patients with the lowest post-surgery PSA nadir and patients who had the lowest PSA at SRT had the fewest deaths. In conclusion, PSA for patients with localized PCa in the pre-PSAR phase of PCa is strongly associated with later PSAR and survival. A rising but still exceedingly low PSA at SRT predicts a good 5-year overall survival.
Collapse
Affiliation(s)
| | - Kalevi Kairemo
- Department of Molecular Radiotherapy & Nuclear Medicine, Docrates Cancer Center, FI-00185 Helsinki, Finland;
| | - Daniel S. Kapp
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
16
|
Koerber SA, Höcht S, Aebersold D, Albrecht C, Boehmer D, Ganswindt U, Schmidt-Hegemann NS, Hölscher T, Mueller AC, Niehoff P, Peeken JC, Pinkawa M, Polat B, Spohn SKB, Wolf F, Zamboglou C, Zips D, Wiegel T. Prostate cancer and elective nodal radiation therapy for cN0 and pN0-a never ending story? : Recommendations from the prostate cancer expert panel of the German Society of Radiation Oncology (DEGRO). Strahlenther Onkol 2024; 200:181-187. [PMID: 38273135 PMCID: PMC10876748 DOI: 10.1007/s00066-023-02193-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024]
Abstract
For prostate cancer, the role of elective nodal irradiation (ENI) for cN0 or pN0 patients has been under discussion for years. Considering the recent publications of randomized controlled trials, the prostate cancer expert panel of the German Society of Radiation Oncology (DEGRO) aimed to discuss and summarize the current literature. Modern trials have been recently published for both treatment-naïve patients (POP-RT trial) and patients after surgery (SPPORT trial). Although there are more reliable data to date, we identified several limitations currently complicating the definitions of general recommendations. For patients with cN0 (conventional or PSMA-PET staging) undergoing definitive radiotherapy, only men with high-risk factors for nodal involvement (e.g., cT3a, GS ≥ 8, PSA ≥ 20 ng/ml) seem to benefit from ENI. For biochemical relapse in the postoperative situation (pN0) and no PSMA imaging, ENI may be added to patients with risk factors according to the SPPORT trial (e.g., GS ≥ 8; PSA > 0.7 ng/ml). If PSMA-PET/CT is negative, ENI may be offered for selected men with high-risk factors as an individual treatment approach.
Collapse
Affiliation(s)
- S A Koerber
- Department of Radiation Oncology, Barmherzige Brüder Hospital Regensburg, Prüfeninger Straße 86, 93049, Regensburg, Germany.
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| | - S Höcht
- Department of Radiation Oncology, Ernst von Bergmann Hospital Potsdam, Charlottenstraße 72, 14467, Potsdam, Germany
| | - D Aebersold
- Department of Radiation Oncology, Inselspital-Bern University Hospital, University of Bern, Freiburgstraße 4, 3010, Bern, Switzerland
| | - C Albrecht
- Nordstrahl Radiation Oncology Unit, Nürnberg North Hospital, Prof.-Ernst-Nathan-Str. 1, 90149, Nürnberg, Germany
| | - D Boehmer
- Department of Radiation Oncology, University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - U Ganswindt
- Department of Radiation Oncology, University Hospital Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - N-S Schmidt-Hegemann
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - T Hölscher
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Fiedlerstraße 19, 01307, Dresden, Germany
| | - A-C Mueller
- Department of Radiation Oncology, RKH Hospital Ludwigsburg, Posilipostraße 4, 71640, Ludwigsburg, Germany
| | - P Niehoff
- Department of Radiation Oncology, Sana Hospital Offenbach, Starkenburgring 66, 63069, Offenbach, Germany
| | - J C Peeken
- Department of Radiation Oncology, Technische Universität München, Ismaninger Straße 22, 81675, Munich, Germany
| | - M Pinkawa
- Department of Radiation Oncology, Robert Janker Klinik, Villenstraße 8, 53129, Bonn, Germany
| | - B Polat
- Department of Radiation Oncology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080, Würzburg, Germany
| | - S K B Spohn
- Department of Radiation Oncology, University Hospital Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
| | - F Wolf
- Department of Radiation Oncology, Paracelsus Medical University of Salzburg, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| | - C Zamboglou
- Department of Radiation Oncology, University Hospital Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
- German Oncology Center, 1, Nikis Avenue, Agios Athanasios, 4108, Limassol, Cyprus
| | - D Zips
- Department of Radiation Oncology, University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - T Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| |
Collapse
|
17
|
Sciarra A, Santarelli V, Salciccia S, Moriconi M, Basile G, Santodirocco L, Carino D, Frisenda M, Di Pierro G, Del Giudice F, Gentilucci A, Bevilacqua G. How the Management of Biochemical Recurrence in Prostate Cancer Will Be Modified by the Concept of Anticipation and Incrementation of Therapy. Cancers (Basel) 2024; 16:764. [PMID: 38398155 PMCID: PMC10886975 DOI: 10.3390/cancers16040764] [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: 01/02/2024] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Biochemical recurrence (BCR) after primary treatments for prostate cancer (PC) is an extremely heterogeneous phase and at least a stratification into low- and high-risk cases for early progression in metastatic disease is necessary. At present, PSA-DT represents the best parameter to define low- and high-risk BCR PC, but real precision medicine is strongly suggested to define tailored management for patients with BCR. Before defining management, it is necessary to exclude the presence of low-volume metastasis associated with PSA progression using new-generation imaging, preferably with PSMA PET/CT. Low-risk BCR cases should be actively observed without early systemic therapies. Early treatment of low-risk BCR with continuous androgen deprivation therapy (ADT) can produce disadvantages such as the development of castration resistance before the appearance of metastases (non-metastatic castration-resistant PC). Patients with high-risk BCR benefit from early systemic therapy. Even with overall survival (OS) as the primary treatment endpoint, metastasis-free survival (MFS) should be used as a surrogate endpoint in clinical trials, especially in long survival stages of the disease. The EMBARK study has greatly influenced the management of high-risk BCR, by introducing the concept of anticipation and intensification through the use of androgen receptor signaling inhibitors (ARSIs) and ADT combination therapy. In high-risk (PSA-DT ≤ 9 months) BCR cases, the combination of enzalutamide with leuprolide significantly improves MFS when compared to leuprolide alone, maintaining an unchanged quality of life in the asymptomatic phase of the disease. The possibility of using ARSIs alone in this early disease setting is suggested by the EMBARK study (arm with enzalutamide alone) with less evidence than with the intensification of the combination therapy. Continued use versus discontinuation of enzalutamide plus leuprolide intensified therapy upon reaching undetectable PSA levels needs to be better defined with further analysis. Real-world analysis must verify the significant results obtained in the context of a phase 3 study.
Collapse
Affiliation(s)
- Alessandro Sciarra
- Department Materno Infantile e Scienze Urologiche, Sapienza University, Viale Policlinico 155, 00161 Rome, Italy; (V.S.); (S.S.); (M.M.); (G.B.); (L.S.); (D.C.); (M.F.); (G.D.P.); (F.D.G.); (A.G.); (G.B.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Efstathiou JA, Morgans AK, Bland CS, Shore ND. Novel hormone therapy and coordination of care in high-risk biochemically recurrent prostate cancer. Cancer Treat Rev 2024; 122:102630. [PMID: 38035646 DOI: 10.1016/j.ctrv.2023.102630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/25/2023] [Indexed: 12/02/2023]
Abstract
Biochemical recurrence (BCR) occurs in 20-50% of patients with prostate cancer (PCa) undergoing primary definitive treatment. Patients with high-risk BCR have an increased risk of metastatic progression and subsequent PCa-specific mortality, and thus could benefit from treatment intensification. Given the increasing complexity of diagnostic and therapeutic modalities, multidisciplinary care (MDC) can play a crucial role in the individualized management of this patient population. This review explores the role for MDC when evaluating the clinical evidence for the evolving definition of high-risk BCR and the emerging therapeutic strategies, especially with novel hormone therapies (NHTs), for patients with either high-risk BCR or oligometastatic PCa. Clinical studies have used different characteristics to define high-risk BCR and there is no consensus regarding the definition of high-risk BCR nor for management strategies. Next-generation imaging and multigene panels offer potential enhanced patient identification and precision-based decision-making, respectively. Treatment intensification with NHTs, either alone or combined with radiotherapy or metastasis-directed therapy, has been promising in clinical trials in patients with high-risk BCR or oligometastases. As novel risk-stratification and treatment options as well as evidence-based literature evolve, it is important to involve a multidisciplinary team to identify patients with high-risk features at an earlier stage, and make informed decisions on the treatments that could optimize their care and long-term outcomes. Nevertheless, MDC data are scarce in the BCR or oligometastatic setting. Efforts to integrate MDC into the standard management of this patient population are needed, and will likely improve outcomes across this heterogeneous PCa patient population.
Collapse
Affiliation(s)
- Jason A Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| | - Alicia K Morgans
- Dana-Farber Cancer Institute, 850 Brookline Ave, Dana 09-930, Boston, MA 02215, USA.
| | - Christopher S Bland
- US Oncology Medical Affairs, Pfizer Inc., 66 Hudson Boulevard, Hudson Yards, Manhattan, New York, NY 10001, USA.
| | - Neal D Shore
- Carolina Urologic Research Center, GenesisCare US, 823 82nd Pkwy, Myrtle Beach, SC, USA.
| |
Collapse
|
19
|
Ah-Thiane L, Sargos P, Chapet O, Jolicoeur M, Terlizzi M, Salembier C, Boustani J, Prevost C, Gaudioz S, Derashodian T, Palumbo S, De Hertogh O, Créhange G, Zilli T, Supiot S. Managing postoperative biochemical relapse in prostate cancer, from the perspective of the Francophone group of Urological radiotherapy (GFRU). Cancer Treat Rev 2023; 120:102626. [PMID: 37734178 DOI: 10.1016/j.ctrv.2023.102626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
Up to 50% of patients treated with radical surgery for localized prostate cancer may experience biochemical recurrence that requires appropriate management. Definitions of biochemical relapse may vary, but, in all cases, consist of an increase in a PSA without clinical or radiological signs of disease. Molecular imaging through to positron emission tomography has taken a preponderant place in relapse diagnosis, progressively replacing bone scan and CT-scan. Prostate bed radiotherapy is currently a key treatment, the action of which should be potentiated by androgen deprivation therapy. Nowadays perspectives consist in determining the best combination therapies, particularly thanks to next-generation hormone therapies, but not exclusively. Several trials are ongoing and should address these issues. We present here a literature review aiming to discuss the current management of biochemical relapse in prostate cancer after radical surgery, in lights of recent findings, as well as future perspectives.
Collapse
Affiliation(s)
- Loic Ah-Thiane
- Department of Radiation Oncology, ICO René Gauducheau, St-Herblain, France
| | - Paul Sargos
- Department of Radiation Oncology, Bergonie Institute, Bordeaux, France
| | - Olivier Chapet
- Department of Radiation Oncology, CHU Lyon Sud, Pierre-Bénite, France
| | - Marjory Jolicoeur
- Department of Radiation Oncology, Charles Le Moyne Hospital, Montreal, Canada
| | - Mario Terlizzi
- Department of Radiation Oncology, Gustave Roussy Cancer Center, Villejuif, France
| | - Carl Salembier
- Department of Radiation Oncology, Europe Hospitals Brussels, Belgium
| | - Jihane Boustani
- Department of Radiation Oncology, CHU Besançon, Besançon, France
| | - Célia Prevost
- Department of Radiation Oncology, CHU Lyon Sud, Pierre-Bénite, France
| | - Sonya Gaudioz
- Department of Radiation Oncology, CHU Lyon Sud, Pierre-Bénite, France
| | - Talar Derashodian
- Department of Radiation Oncology, Sindi Ahluwalia Hawkins Centre, Kelowna, Canada
| | - Samuel Palumbo
- Department of Radiation Oncology, CHU UCL Namur-Sainte Elisabeth, Namur, Belgium
| | - Olivier De Hertogh
- Department of Radiation Oncology, CHR Verviers East Belgium, Verviers, Belgium
| | - Gilles Créhange
- Department of Radiation Oncology, Curie Institute, Saint-Cloud, France
| | - Thomas Zilli
- Department of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Stéphane Supiot
- Department of Radiation Oncology, ICO René Gauducheau, St-Herblain, France.
| |
Collapse
|
20
|
Argalácsová S, Vočka M, Čapoun O, Lambert L. Timing of Early Salvage Therapy for Patients With Biochemical Relapse of Prostate Carcinoma. Oncol Rev 2023; 17:10676. [PMID: 37771544 PMCID: PMC10522833 DOI: 10.3389/or.2023.10676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/30/2023] [Indexed: 09/30/2023] Open
Abstract
Between 25% and 33% of patients after radical prostatectomy experience a relapse of the disease. The risk of relapse increases in patients with risk factors up to 50%-80%. For a long time, adjuvant radiotherapy has been considered the standard of care. Four large prospective trials, that compared adjuvant and salvage radiotherapy in patients with biochemical relapse, showed the superiority of the adjuvant approach in biochemical and local relapse-free survival, but no consistent benefit in long-term endpoints (i.e., metastasis-free survival, overall survival, or carcinoma-specific survival) at the expense of increased urinary and bowel toxicity. Three large international studies comparing adjuvant and salvage radiotherapy paved the way toward early salvage radiotherapy. However, the optimal threshold of the PSA level (range of 0.2-0.5 ng/mL) for initiating early salvage radiotherapy remains unresolved and still poses a challenge in everyday clinical practice when balancing the need for early radiotherapy and the associated toxicity. Imprecise stratification of biochemical relaps patients according to the risk of clinical relapse drives efforts to find additional molecular biomarkers that would improve the timing of the salvage therapy.
Collapse
Affiliation(s)
- Soňa Argalácsová
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Michal Vočka
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Otakar Čapoun
- Department of Urology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Lukáš Lambert
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| |
Collapse
|
21
|
Dao J, Conway PJ, Subramani B, Meyyappan D, Russell S, Mahadevan D. Using cfDNA and ctDNA as Oncologic Markers: A Path to Clinical Validation. Int J Mol Sci 2023; 24:13219. [PMID: 37686024 PMCID: PMC10487653 DOI: 10.3390/ijms241713219] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The detection of circulating tumor DNA (ctDNA) in liquid biopsy samples as an oncological marker is being used in clinical trials at every step of clinical management. As ctDNA-based liquid biopsy kits are developed and used in clinics, companies work towards increased convenience, accuracy, and cost over solid biopsies and other oncological markers. The technology used to differentiate ctDNA and cell-free DNA (cfDNA) continues to improve with new tests and methodologies being able to detect down to mutant allele frequencies of 0.001% or 1/100,000 copies. Recognizing this development in technology, the FDA has recently given pre-market approval and breakthrough device designations to multiple companies. The purpose of this review is to look at the utility of measuring total cfDNA, techniques used to differentiate ctDNA from cfDNA, and the utility of different ctDNA-based liquid biopsy kits using relevant articles from PubMed, clinicaltrials.gov, FDA approvals, and company newsletters. Measuring total cfDNA could be a cost-effective, viable prognostic marker, but various factors do not favor it as a monitoring tool during chemotherapy. While there may be a place in the clinic for measuring total cfDNA in the future, the lack of standardization means that it is difficult to move forward with large-scale clinical validation studies currently. While the detection of ctDNA has promising standardized liquid biopsy kits from various companies with large clinical trials ongoing, their applications in screening and minimal residual disease can suffer from lower sensitivity. However, researchers are working towards solutions to these issues with innovations in technology, multi-omics, and sampling. With great promise, further research is needed before liquid biopsies can be recommended for everyday clinical management.
Collapse
Affiliation(s)
- Jonathan Dao
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Patrick J. Conway
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Baskaran Subramani
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Devi Meyyappan
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
| | - Sammy Russell
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Daruka Mahadevan
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
22
|
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
|
23
|
Sutera P, Skinner H, Witek M, Mishra M, Kwok Y, Davicioni E, Feng F, Song D, Nichols E, Tran PT, Bergom C. Histology Specific Molecular Biomarkers: Ushering in a New Era of Precision Radiation Oncology. Semin Radiat Oncol 2023; 33:232-242. [PMID: 37331778 PMCID: PMC10446901 DOI: 10.1016/j.semradonc.2023.03.001] [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] [Indexed: 06/20/2023]
Abstract
Histopathology and clinical staging have historically formed the backbone for allocation of treatment decisions in oncology. Although this has provided an extremely practical and fruitful approach for decades, it has long been evident that these data alone do not adequately capture the heterogeneity and breadth of disease trajectories experienced by patients. As efficient and affordable DNA and RNA sequencing have become available, the ability to provide precision therapy has become within grasp. This has been realized with systemic oncologic therapy, as targeted therapies have demonstrated immense promise for subsets of patients with oncogene-driver mutations. Further, several studies have evaluated predictive biomarkers for response to systemic therapy within a variety of malignancies. Within radiation oncology, the use of genomics/transcriptomics to guide the use, dose, and fractionation of radiation therapy is rapidly evolving but still in its infancy. The genomic adjusted radiation dose/radiation sensitivity index is one such early and exciting effort to provide genomically guided radiation dosing with a pan-cancer approach. In addition to this broad method, a histology specific approach to precision radiation therapy is also underway. Herein we review select literature surrounding the use of histology specific, molecular biomarkers to allow for precision radiotherapy with the greatest emphasis on commercially available and prospectively validated biomarkers.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Heath Skinner
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew Witek
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark Mishra
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Young Kwok
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Felix Feng
- Departments of Radiation Oncology, Medicine and Urology, UCSF, San Francisco, CA, USA
| | - Daniel Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth Nichols
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Phuoc T. Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
24
|
Herberts C, Wyatt AW, Nguyen PL, Cheng HH. Genetic and Genomic Testing for Prostate Cancer: Beyond DNA Repair. Am Soc Clin Oncol Educ Book 2023; 43:e390384. [PMID: 37207301 DOI: 10.1200/edbk_390384] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Significant progress has been made in genetic and genomic testing for prostate cancer across the disease spectrum. Molecular profiling is increasingly relevant for routine clinical management, fueled in part by advancements in testing technology and integration of biomarkers into clinical trials. In metastatic prostate cancer, defects in DNA damage response genes are now established predictors of benefit to US Food and Drug Administration-approved poly (ADP-ribose) polymerase inhibitors and immune checkpoint inhibitors, and trials are actively investigating these and other targeted treatment strategies in earlier disease states. Excitingly, opportunities for molecularly informed management beyond DNA damage response genes are also maturing. Germline genetic variants (eg, BRCA2 or MSH2/6) and polygenic germline risk scores are being investigated to inform cancer screening and active surveillance in at-risk carriers. RNA expression tests have recently gained traction in localized prostate cancer, enabling patient risk stratification and tailored treatment intensification via radiotherapy and/or androgen deprivation therapy for localized or salvage treatment. Finally, emerging minimally invasive circulating tumor DNA technology promises to enhance biomarker testing in advanced disease pending additional methodological and clinical validation. Collectively, genetic and genomic tests are rapidly becoming indispensable tools for informing the optimal clinical management of prostate cancer.
Collapse
Affiliation(s)
- Cameron Herberts
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Paul L Nguyen
- Harvard Medical School, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA
| | - Heather H Cheng
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
25
|
Latorzeff I, Le Guevelou J, Sargos P. Radiation therapy post radical prostatectomy: who, when and why? Curr Opin Support Palliat Care 2023; 17:47-54. [PMID: 36367534 DOI: 10.1097/spc.0000000000000627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE OF REVIEW During decades, adjuvant radiotherapy (ART) has been the standard of care after surgery, based on four randomized clinical trials (RCTs). As early salvage radiotherapy (SRT) recently challenged the ART paradigm, the optimal timing to initiate radiotherapy remains a matter of debate. RECENT FINDINGS Three RCTs evaluated ART or SRT for postprostatectomy patients, with pathological risk factors (Gleason score > 8, pT3, positive margins). The ARTISTIC meta-analysis demonstrated similar 5-year biochemical recurrence-free survival for ART and SRT (89 vs. 88%). Lower rates of late genitourinary toxicity were demonstrated within the SRT arm, favouring early SRT in clinical practice.The addition of pelvic lymph node radiotherapy recently demonstrated an improvement in freedom from progression within the randomized RTOG 0534 trial, especially for patients with pretreatment prostate serum antigen (PSA) levels more than 0.35 ng/ml. The most appropriate androgen deprivation therapy duration remains a point of controversy. SUMMARY The SRT approach can be favoured for the vast majority of patients, provided close monitoring of PSA and early treatment in the event of biochemical recurrence. Radiotherapy dose and volumes and ADT use might be correlated to SRT timing (early vs. late). Results from trials including genomic tests and metabolic imaging will probably help to refine these criteria.
Collapse
Affiliation(s)
- Igor Latorzeff
- Department of Radiation Oncology, Clinique Pasteur, Toulouse, France
| | - Jennifer Le Guevelou
- Department of Radiation Oncology, Geneva University Hospital, and Faculty of Medicine, Geneva, Switzerland
| | - Paul Sargos
- Department of Radiation Oncology, Institut Bergonié, Bordeaux, France
| |
Collapse
|
26
|
Gillessen S, Bossi A, Davis ID, de Bono J, Fizazi K, James ND, Mottet N, Shore N, Small E, Smith M, Sweeney C, Tombal B, Antonarakis ES, Aparicio AM, Armstrong AJ, Attard G, Beer TM, Beltran H, Bjartell A, Blanchard P, Briganti A, Bristow RG, Bulbul M, Caffo O, Castellano D, Castro E, Cheng HH, Chi KN, Chowdhury S, Clarke CS, Clarke N, Daugaard G, De Santis M, Duran I, Eeles R, Efstathiou E, Efstathiou J, Ngozi Ekeke O, Evans CP, Fanti S, Feng FY, Fonteyne V, Fossati N, Frydenberg M, George D, Gleave M, Gravis G, Halabi S, Heinrich D, Herrmann K, Higano C, Hofman MS, Horvath LG, Hussain M, Jereczek-Fossa BA, Jones R, Kanesvaran R, Kellokumpu-Lehtinen PL, Khauli RB, Klotz L, Kramer G, Leibowitz R, Logothetis CJ, Mahal BA, Maluf F, Mateo J, Matheson D, Mehra N, Merseburger A, Morgans AK, Morris MJ, Mrabti H, Mukherji D, Murphy DG, Murthy V, Nguyen PL, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Pezaro C, Poon DMC, Pritchard CC, Rabah DM, Rathkopf D, Reiter RE, Rubin MA, Ryan CJ, Saad F, Pablo Sade J, Sartor OA, Scher HI, Sharifi N, Skoneczna I, Soule H, Spratt DE, Srinivas S, Sternberg CN, Steuber T, Suzuki H, Sydes MR, Taplin ME, Tilki D, Türkeri L, Turco F, Uemura H, Uemura H, Ürün Y, Vale CL, van Oort I, Vapiwala N, Walz J, Yamoah K, Ye D, Yu EY, Zapatero A, Zilli T, Omlin A. Management of Patients with Advanced Prostate Cancer. Part I: Intermediate-/High-risk and Locally Advanced Disease, Biochemical Relapse, and Side Effects of Hormonal Treatment: Report of the Advanced Prostate Cancer Consensus Conference 2022. Eur Urol 2023; 83:267-293. [PMID: 36494221 PMCID: PMC7614721 DOI: 10.1016/j.eururo.2022.11.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Innovations in imaging and molecular characterisation and the evolution of new therapies have improved outcomes in advanced prostate cancer. Nonetheless, we continue to lack high-level evidence on a variety of clinical topics that greatly impact daily practice. To supplement evidence-based guidelines, the 2022 Advanced Prostate Cancer Consensus Conference (APCCC 2022) surveyed experts about key dilemmas in clinical management. OBJECTIVE To present consensus voting results for select questions from APCCC 2022. DESIGN, SETTING, AND PARTICIPANTS Before the conference, a panel of 117 international prostate cancer experts used a modified Delphi process to develop 198 multiple-choice consensus questions on (1) intermediate- and high-risk and locally advanced prostate cancer, (2) biochemical recurrence after local treatment, (3) side effects from hormonal therapies, (4) metastatic hormone-sensitive prostate cancer, (5) nonmetastatic castration-resistant prostate cancer, (6) metastatic castration-resistant prostate cancer, and (7) oligometastatic and oligoprogressive prostate cancer. Before the conference, these questions were administered via a web-based survey to the 105 physician panel members ("panellists") who directly engage in prostate cancer treatment decision-making. Herein, we present results for the 82 questions on topics 1-3. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Consensus was defined as ≥75% agreement, with strong consensus defined as ≥90% agreement. RESULTS AND LIMITATIONS The voting results reveal varying degrees of consensus, as is discussed in this article and shown in the detailed results in the Supplementary material. The findings reflect the opinions of an international panel of experts and did not incorporate a formal literature review and meta-analysis. CONCLUSIONS These voting results by a panel of international experts in advanced prostate cancer 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 prioritise areas for future research. Diagnostic and treatment decisions should always be individualised based on patient and cancer characteristics (disease extent and location, treatment history, comorbidities, and patient preferences) and should incorporate current and emerging clinical evidence, therapeutic guidelines, and logistic and economic factors. Enrolment in clinical trials is always strongly encouraged. Importantly, APCCC 2022 once again identified important gaps (areas of nonconsensus) that merit evaluation in specifically designed trials. PATIENT SUMMARY The Advanced Prostate Cancer Consensus Conference (APCCC) provides a forum to discuss and debate current diagnostic and treatment options for patients with advanced prostate cancer. The conference aims to share the knowledge of international experts in prostate cancer with health care providers and patients worldwide. At each APCCC, a panel of physician experts vote in response to multiple-choice questions about their clinical opinions and approaches to managing advanced prostate cancer. This report presents voting results for the subset of questions pertaining to intermediate- and high-risk and locally advanced prostate cancer, biochemical relapse after definitive treatment, advanced (next-generation) imaging, and management of side effects caused by hormonal therapies. The results provide a practical guide to help clinicians and patients discuss treatment options as part of shared multidisciplinary decision-making. The findings may be especially useful when there is little or no high-level evidence to guide treatment decisions.
Collapse
Affiliation(s)
- Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland.
| | - Alberto Bossi
- Genitourinary Oncology, Prostate Brachytherapy Unit, Gustave Roussy, Paris, France
| | - Ian D Davis
- Monash University and Eastern Health, Victoria, Australia
| | - Johann de Bono
- The Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | | | | | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA; Urology/Surgical Oncology, GenesisCare, Myrtle Beach, SC, USA
| | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Mathew Smith
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Christopher Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Ana M Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
| | | | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Himisha Beltran
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anders Bjartell
- Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Pierre Blanchard
- Département de Radiothérapie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, URI, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Rob G Bristow
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Christie NHS Trust and CRUK Manchester Institute and Cancer Centre, Manchester, UK
| | - Muhammad Bulbul
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | - Daniel Castellano
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elena Castro
- Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain
| | - Heather H Cheng
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Kim N Chi
- BC Cancer, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Caroline S Clarke
- Research Department of Primary Care & Population Health, Royal Free Campus, University College London, London, UK
| | - Noel Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - Gedske Daugaard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Ignacio Duran
- Department of Medical Oncology, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Cantabria, Spain
| | - Ros Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | | | - Jason Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Onyeanunam Ngozi Ekeke
- Department of Surgery, University of Port Harcourt Teaching Hospital, Alakahia, Port Harcourt, Nigeria
| | | | - Stefano Fanti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Felix Y Feng
- University of California San Francisco, San Francisco, CA, USA
| | - Valerie Fonteyne
- Department of Radiation-Oncology, Ghent University Hospital, Ghent, Belgium
| | - Nicola Fossati
- Department of Urology, Ospedale Regionale di Lugano, Civico USI - Università della Svizzera Italiana, Lugano, Switzerland
| | - Mark Frydenberg
- Department of Surgery, Prostate Cancer Research Program, Monash University, Melbourne, Australia; Department of Anatomy & Developmental Biology, Faculty of Nursing, Medicine & Health Sciences, Monash University, Melbourne, Australia
| | - Daniel George
- Department of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA; Department of Surgery, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Martin Gleave
- Urological Sciences, Vancouver Prostate Centre, 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 and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Celestia Higano
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Lisa G Horvath
- Chris O'Brien Lifehouse, Camperdown, NSW, Australia; Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia
| | - Maha 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 Radiotherapy, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Robert Jones
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Pirkko-Liisa Kellokumpu-Lehtinen
- Faculty of Medicine and Health Technology, Tampere University and Tampere Cancer Center, Tampere, Finland; Research, Development and Innovation Center, Tampere University Hospital, Tampere, Finland
| | - Raja B Khauli
- Department of Urology and the Naef K. Basile Cancer Institute (NKBCI), American University of Beirut Medical Center, Beirut, Lebanon
| | - Laurence Klotz
- Division of Urology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Raya Leibowitz
- Oncology Institute, Shamir Medical Center, Be'er Ya'akov, Israel; Faculty of Medicine, Tel-Aviv University, Israel
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; University of Athens Alexandra Hospital, Athens, Greece
| | - Brandon A Mahal
- Department of Radiation Oncology, University of Miami Sylvester Cancer Center, Miami, FL, USA
| | - Fernando Maluf
- Beneficiência Portuguesa de São Paulo, São Paulo, SP, Brasil; Departamento de Oncologia, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Joaquin Mateo
- Department of Medical Oncology and Prostate Cancer Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO) and Vall d'Hebron University Hospital, Barcelona, Spain
| | - David Matheson
- Faculty of Education, Health and Wellbeing, Walsall Campus, Walsall, UK
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Axel Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Alicia K Morgans
- Dana-Farber Cancer Institute and 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
- National Institute of Oncology, Mohamed V University, Rabat, Morocco
| | - Deborah Mukherji
- Clemenceau Medical Center, Dubai, United Arab Emirates; Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | | | - 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, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Netwerk, 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, Northern Ireland Cancer Centre, Belfast City Hospital, Belfast, Northern Ireland
| | - Anwar R Padhani
- Mount Vernon Cancer Centre and Institute of Cancer Research, London, UK
| | - Carmel Pezaro
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Darren M C Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong; The Chinese University of Hong Kong, Shatin, Hong Kong
| | - 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, KFSHRC, Riyadh, Saudi Arabia
| | - Dana Rathkopf
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Mark A Rubin
- Bern Center for Precision Medicine and Department for Biomedical Research, Bern, Switzerland
| | - Charles J Ryan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Fred Saad
- Centre Hospitalier de Université de Montréal, Montreal, Quebec, Canada
| | | | | | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Nima Sharifi
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA; Department of Cancer Biology, GU Malignancies Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Iwona Skoneczna
- Rafal Masztak Grochowski Hospital, Maria Sklodowska Curie National Research Institute of Oncology, Warsaw, Poland
| | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | - Daniel E Spratt
- 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
| | - Thomas Steuber
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Levent Türkeri
- Department of Urology, M.A. Aydınlar Acıbadem University, Altunizade Hospital, Istanbul, Turkey
| | - Fabio Turco
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Hiroji Uemura
- Yokohama City University Medical Center, Yokohama, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University School of Medicine, Ankara, Turkey; Ankara University Cancer Research Institute, Ankara, Turkey
| | - Claire L Vale
- University College London, MRC Clinical Trials Unit at UCL, London, UK
| | - Inge van Oort
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Neha Vapiwala
- Department of Radiation Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jochen Walz
- Department of Urology, Institut Paoli-Calmettes Cancer Centre, Marseille, France
| | - Kosj Yamoah
- Department of Radiation Oncology & Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, 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 Oncology, University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Almudena Zapatero
- Department of Radiation Oncology, Hospital Universitario de La Princesa, Health Research Institute, Madrid, Spain
| | - Thomas Zilli
- Radiation Oncology, Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aurelius Omlin
- Onkozentrum Zurich, University of Zurich and Tumorzentrum Hirslanden Zurich, Switzerland
| |
Collapse
|
27
|
Valle LF, Jiang T, Weiner AB, Reiter RE, Rettig MB, Shen J, Chang AJ, Nickols NG, Steinberg ML, Kishan AU. Multimodality Therapies for Localized Prostate Cancer. Curr Oncol Rep 2023; 25:221-229. [PMID: 36723856 PMCID: PMC11288626 DOI: 10.1007/s11912-023-01374-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW Multimodality therapy including radical prostatectomy, radiation therapy, and hormone therapy are frequently deployed in the management of localized prostate cancer. We sought to perform a critical appraisal of the most contemporary literature focusing on the multimodality management of localized prostate cancer. RECENT FINDINGS Men who are ideal candidates for multimodality therapy include those with unfavorable intermediate-risk disease, high-risk disease, and very high-risk disease. Enhancements in both systemic agents (including second-generation antiandrogens) as well as localized therapies (such as stereotactic body radiotherapy and brachytherapy) are refining the optimal balance between the use of systemic and local therapies for localized prostate cancer. Genomic predictors are emerging as critical tools for more precisely allocating treatment intensification with multimodality therapies as well as treatment de-intensification. Close collaboration among medical oncologists, surgeons, and radiation oncologists will be critical for coordinating evidence-based multimodality therapies when clearly indicated and for supporting shared decision-making in areas where the evidence is mixed.
Collapse
Affiliation(s)
- Luca F Valle
- Department of Radiation Oncology, University of California Los Angeles, 200 Medical Plaza, Suite B265, Los Angeles, CA, 90095, USA
- Department of Radiation Oncology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, USA
| | - Tommy Jiang
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, USA
| | - Adam B Weiner
- Department of Urology, University of California Los Angeles, Los Angeles, USA
| | - Robert E Reiter
- Department of Urology, University of California Los Angeles, Los Angeles, USA
| | - Matthew B Rettig
- Department of Hematology/Oncology, University of California Los Angeles, Los Angeles, USA
- Department of Hematology/Oncology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, USA
| | - John Shen
- Department of Hematology/Oncology, University of California Los Angeles, Los Angeles, USA
| | - Albert J Chang
- Department of Radiation Oncology, University of California Los Angeles, 200 Medical Plaza, Suite B265, Los Angeles, CA, 90095, USA
| | - Nicholas G Nickols
- Department of Radiation Oncology, University of California Los Angeles, 200 Medical Plaza, Suite B265, Los Angeles, CA, 90095, USA
- Department of Radiation Oncology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, USA
- Department of Urology, University of California Los Angeles, Los Angeles, USA
| | - Michael L Steinberg
- Department of Radiation Oncology, University of California Los Angeles, 200 Medical Plaza, Suite B265, Los Angeles, CA, 90095, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, 200 Medical Plaza, Suite B265, Los Angeles, CA, 90095, USA.
- Department of Urology, University of California Los Angeles, Los Angeles, USA.
| |
Collapse
|
28
|
ESTRO-ACROP recommendations for evidence-based use of androgen deprivation therapy in combination with external-beam radiotherapy in prostate cancer. Radiother Oncol 2023; 183:109544. [PMID: 36813168 DOI: 10.1016/j.radonc.2023.109544] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND AND PURPOSE There is no consensus concerning the appropriate use of androgen deprivation therapy (ADT) during primary and postoperative external-beam radiotherapy (EBRT) in the management of prostate cancer (PCa). Thus, the European Society for Radiotherapy and Oncology (ESTRO) Advisory Committee for Radiation Oncology Practice (ACROP) guidelines seeks to present current recommendations for the clinical use of ADT in the various indications of EBRT. MATERIAL AND METHODS A literature search was conducted in MEDLINE PubMed that evaluated EBRT and ADT in prostate cancer. The search focused on randomized, Phase II and III trials published in English from January 2000 to May 2022. In case topics were addressed in the absence of Phase II or III trials, recommendations were labelled accordingly based on the limited body of evidence. Localized PCa was classified according to D'Amico et al. classification in low-, intermediate and high risk PCa. The ACROP clinical committee identified 13 European experts who discussed and analyzed the body of evidence concerning the use of ADT with EBRT for prostate cancer. RESULTS Key issues were identified and are discussed: It was concluded that no additional ADT is recommended for low-risk prostate cancer patients, whereas for intermediate- and high-risk patients four to six months and two to three years of ADT are recommended. Likewise, patients with locally advanced prostate cancer are recommended to receive ADT for two to three years and when ≥ 2 high-risk factors (cT3-4, ISUP grade ≥ 4 or PSA ≥ 40 ng/ml) or cN1 is present ADT for three years plus additional Abiraterone for two years is recommended. For postoperative patients no ADT is recommended for adjuvant EBRT in pN0 patients whereas for pN1 patients adjuvant EBRT with long-term ADT is performed for at least 24 to 36 months. In the setting of salvage EBRT ADT is performed in biochemically persistent PCa patients with no evidence of metastatic disease. Long-term ADT (24 months) is recommended in pN0 patients with high risk of further progression (PSA ≥ 0.7 ng/ml and ISUP grade group ≥ 4) and a life expectancy of over ten years, whereas short-term ADT (6 months) is recommended in pN0 patients with lower risk profile (PSA < 0.7 ng/ml and ISUP grade group 4). Patients considered for ultra-hypofractionated EBRT as well as patients with image based local recurrence within the prostatic fossa or lymph node recurrence should participate in appropriate clinical trials evaluating the role of additional ADT. CONCLUSION These ESTRO-ACROP recommendations are evidence-based and relevant to the use of ADT in combination with EBRT in PCa for the most common clinical settings.
Collapse
|
29
|
Tran PT, Lowe K, Tsai HL, Song DY, Hung AY, Hearn JW, Miller S, Proudfoot JA, Deek MP, Phillips R, Lotan T, Paller CJ, Marshall CH, Markowski M, Dipasquale S, Denmeade S, Carducci M, Eisenberger M, DeWeese TL, Orton M, Deville C, Davicioni E, Liauw SL, Heath EI, Greco S, Desai NB, Spratt DE, Feng F, Wang H, Beer TM, Antonarakis ES. Phase II Randomized Study of Salvage Radiation Therapy Plus Enzalutamide or Placebo for High-Risk Prostate-Specific Antigen Recurrent Prostate Cancer After Radical Prostatectomy: The SALV-ENZA Trial. J Clin Oncol 2023; 41:1307-1317. [PMID: 36367998 PMCID: PMC9940936 DOI: 10.1200/jco.22.01662] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE We sought to investigate whether enzalutamide (ENZA), without concurrent androgen deprivation therapy, increases freedom from prostate-specific antigen (PSA) progression (FFPP) when combined with salvage radiation therapy (SRT) in men with recurrent prostate cancer after radical prostatectomy (RP). PATIENTS AND METHODS Men with biochemically recurrent prostate cancer after RP were enrolled into a randomized, double-blind, phase II, placebo-controlled, multicenter study of SRT plus ENZA or placebo (ClinicalTrials.gov identifier: NCT02203695). Random assignment (1:1) was stratified by center, surgical margin status (R0 v R1), PSA before salvage treatment (PSA ≥ 0.5 v < 0.5 ng/mL), and pathologic Gleason sum (7 v 8-10). Patients were assigned to receive either ENZA 160 mg once daily or matching placebo for 6 months. After 2 months of study drug therapy, external-beam radiation (66.6-70.2 Gy) was administered to the prostate bed (no pelvic nodes). The primary end point was FFPP in the intention-to-treat population. Secondary end points were time to local recurrence within the radiation field, metastasis-free survival, and safety as determined by frequency and severity of adverse events. RESULTS Eighty-six (86) patients were randomly assigned, with a median follow-up of 34 (range, 0-52) months. Trial arms were well balanced. The median pre-SRT PSA was 0.3 (range, 0.06-4.6) ng/mL, 56 of 86 patients (65%) had extraprostatic disease (pT3), 39 of 86 (45%) had a Gleason sum of 8-10, and 43 of 86 (50%) had positive surgical margins (R1). FFPP was significantly improved with ENZA versus placebo (hazard ratio [HR], 0.42; 95% CI, 0.19 to 0.92; P = .031), and 2-year FFPP was 84% versus 66%, respectively. Subgroup analyses demonstrated differential benefit of ENZA in men with pT3 (HR, 0.22; 95% CI, 0.07 to 0.69) versus pT2 disease (HR, 1.54; 95% CI, 0.43 to 5.47; Pinteraction = .019) and R1 (HR, 0.14; 95% CI, 0.03 to 0.64) versus R0 disease (HR, 1.00; 95% CI, 0.36 to 2.76; Pinteraction = .023). There were insufficient secondary end point events for analysis. The most common adverse events were grade 1-2 fatigue (65% ENZA v 53% placebo) and urinary frequency (40% ENZA v 49% placebo). CONCLUSION SRT plus ENZA monotherapy for 6 months in men with PSA-recurrent high-risk prostate cancer after RP is safe and delays PSA progression relative to SRT alone. The impact of ENZA on distant metastasis or survival is unknown at this time.
Collapse
Affiliation(s)
- Phuoc T. Tran
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Current address: Department of Radiation Oncology, University of Maryland, Baltimore, MD
| | - Kathryn Lowe
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Hua-Ling Tsai
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Daniel Y. Song
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Arthur Y. Hung
- Department of Radiation Medicine, OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Jason W.D. Hearn
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Steven Miller
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI
| | | | - Matthew P. Deek
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ryan Phillips
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tamara Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Channing J. Paller
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Catherine H. Marshall
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mark Markowski
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shirl Dipasquale
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Samuel Denmeade
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michael Carducci
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mario Eisenberger
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Theodore L. DeWeese
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Matthew Orton
- Department of Radiation Oncology, Indiana University Health Arnett, Lafayette, IN
| | - Curtiland Deville
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Stanley L. Liauw
- Department of Radiation Oncology and Cellular Oncology, University of Chicago, Chicago, IL
| | - Elisabeth I. Heath
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI
| | - Stephen Greco
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Neil B. Desai
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Daniel E. Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Felix Feng
- Departments of Medicine, Radiation Oncology and Urology, University of California San Francisco, San Francisco, CA
| | - Hao Wang
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tomasz M. Beer
- OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Emmanuel S. Antonarakis
- Department of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medicine, University of Minnesota, Minneapolis, MN
| |
Collapse
|
30
|
Lee EE, Singh T, Hu C, Han M, Deville CJ, Halthore A, Greco S, Tran P, DeWeese T, Song DY. The impact of salvage radiotherapy initiation at PSA ≤ 0.5 ng/ml on metastasis-free survival in patients with relapsed prostate cancer following prostatectomy. Prostate 2023; 83:190-197. [PMID: 36316967 DOI: 10.1002/pros.24452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Salvage radiation therapy (SRT) is indicated for biochemical failure after radical prostatectomy. Prior data have shown that initiation of SRT at lower PSA levels improves subsequent biochemical control, yet given the long natural history of prostate cancer questions remain regarding optimal timing of SRT. We analyzed the impact of prostate specific antigen (PSA) level at time of salvage radiotherapy with regard to both biochemical relapse-free (bRFS) as well as metastasis-free survival (MFS) in patients with biochemically recurrent prostate cancer. METHODS Using prospective institutional tumor registry data, univariate and multivariable-adjusted Cox proportional hazards models were constructed to assess association between outcomes and clinical and pathologic prognostic features, including pre-SRT PSA, interval from prostatectomy to SRT, androgen deprivation therapy (ADT), and adverse pathologic features. RESULTS We identified 397 patients who received salvage RT between 1985 and 2016: 187 (45.8%) received SRT initiated when pre-RT PSA was ≤0.5 ng/ml; 212 (52.0%) patients had pre-SRT PSA > 0.5 ng/ml. Independent of pathologic risk status and ADT use, pre-SRT PSA ≤ 0.5 ng/ml was the most significant predictor of bRFS (HR 0.39, 95% CI [0.27, 0.56]) as well as MFS (HR = 0.58, 95% CI [0.37, 0.91]). Seminal vesicle invasion was also associated with shorter interval to biochemical failure, HR = 1.79, 95% CI [1.07, 2.98], and eventual metastases, HR = 2.07, 95% CI [1.14, 3.740]. CONCLUSIONS Initiation of salvage RT while PSA levels remain ≤0.5 ng/ml was associated with improved MFS. Consideration for salvage RT initiation while PSA levels remain low is warranted to minimize risk of future prostate cancer metastasis.
Collapse
Affiliation(s)
- Emerson E Lee
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tanmay Singh
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Hu
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Misop Han
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Curtiland Jr Deville
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aditya Halthore
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stephen Greco
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Phuoc Tran
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Theodore DeWeese
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel Y Song
- Department of Radiation Oncology & Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
31
|
Liang Z, Lin S, Lai H, Li L, Wu J, Zhang H, Fang C. Efficacy and safety of salvage radiotherapy combined with endocrine therapy in patients with biochemical recurrence after radical prostatectomy: A systematic review and meta-analysis of randomized controlled trials. Front Oncol 2023; 12:1093759. [PMID: 36761425 PMCID: PMC9902708 DOI: 10.3389/fonc.2022.1093759] [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: 11/09/2022] [Accepted: 12/30/2022] [Indexed: 01/25/2023] Open
Abstract
Background The addition of endocrine therapy to salvage radiotherapy (SRT) is expected to further improve the prognosis of patients with biochemical recurrence of prostate cancer after radical prostatectomy (RP). The quantitative synthesis of clinical outcomes of SRT combined with endocrine therapy is limited. Whether salvage radiotherapy plus endocrine therapy remains inconclusive. We performed a systematic review and meta-analysis of existing randomized controlled trials to evaluate the efficacy and safety of salvage radiotherapy combined with endocrine therapy in patients with biochemical recurrence after radical prostatectomy. Methods A systematic search of PubMed, EMBASE, and the Cochrane library was performed for articles published between January 1, 2012 and October 10, 2022. Data were analyzed using Review Manager 5.4.1 (Cochrane Collaboration Software). Main outcome and measures included biochemical progression-free survival (bPFS), metastasis free survival (MFS), overall survival (OS), and Grade 3 or higher adverse events (3+AEs), including acute and late adverse events. Results In this systematic review and meta-analysis, 4 randomized controlled studies enrolling 2731 male (1374 of whom received SRT combined with endocrine therapy and 1357 controls) met the inclusion criteria. SRT combined with endocrine therapy were related to significantly improve bPFS (HR=0.52; 95% CI: 0.46 0.59; p<0.00001) and MFS (HR=0.75; 95% CI: 0.64 0.88; p<0.001). Compared with SRT alone, the combination therapy tended to be associated with prolong OS (HR=0.83; 95% CI: 0.69-1.01; p=0.06), but not statistically significant. At early follow-up, the risk of acute AEs was comparable in the two groups (RR=1.04; 95% CI: 0.22-4.85). However, the risk of late AEs was higher in the combination group at later follow-up (RR=1.33; 95% CI: 1.09-1.62). Conclusions This systematic review and meta-analysis found superior efficacy associated with adding endocrine therapy to SRT compared with SRT alone in patients with biochemical recurrence after RP. Additional endocrine therapy is safe and feasible for patients with biochemical recurrence after RP. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier (CRD42022365432).
Collapse
|
32
|
Al-Nasralla ASH, Hussian SS, Tektook NK. Immunological analysis of Interleukin-10 (IL-10), tumor necrosis factor-a (TNF-a), and Prostate-specific antigen (PSA) in benign and malignant prostate cancer. Hum Antibodies 2023:HAB220018. [PMID: 36776045 DOI: 10.3233/hab-220018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
INTRODUCTION Among the cancers that impacts men, prostate cancer considerably raises deaths for males around the world. Persons with tumours can have a localized or advanced form of the illness. OBJECTIVE The present study aimed to determining the relationship between the level of cytokines (IL-10 and TNF-a) and PSA in the sera of patients and compared it with healthy. MATERIALS AND METHODS A case control study consist of three group included was in this study. The first group involves 50 patients with PC were observation in Al-Amal Oncology Hospital in the period from April 2021 to April 2022 under the supervision of oncology specialists was included in this study. Second group consist of 30 patients. They have benign hyper plaisa (BHP), this group has been collected from urosergical department . Third group was include 20 healthy volunteers (non prostate cancer and non BHP). Prostate specific antigen (PSA) was measured by mini - VIDAS device using kit supplied by Biomerieux - France. IL-10 and TNF-a levels were measured by ELISA technique using kit supplied by CAUSABIO - China. RESULTS Results of the present study showed the 60-69 years age group scored highest percentage in benign (56.7%), malignant (54.0%), compared to control (healthy) (50.0%), while > 69 years scored least percentage in these groups (3.3%, 14.0%, and 25.0%) respectively with significant different (p< 0.05). Additionally, the IL-10 and PSA scored highest mean levels in the malignant group (1.22 ± 0.23 and 27.66 ± 6.31), while TNF-a scored highest mean levels in a benign group (0.30 ± 0.11). The least mean level of IL-10 was in healthy (0.42 ± 0.15), TNF-a in malignant (0.23 ± 0.03), and PSA in benign (6.73 ± 1.36). Finally, there is a significant difference among age groups and PSA, IL-10, and TNF-parameters. CONCLUSIONS We concluded the PSA, TNF-a and IL-10 parameters are play important roles in pathogenesis patients with prostate cancer. PCa is high prevalence in elderly population.
Collapse
|
33
|
Re: The Addition of Androgen Deprivation Therapy and Pelvic Lymph Node Treatment to Prostate Bed Salvage Radiotherapy (NRG Oncology/RTOG 0534 SPPORT): An International, Multicentre, Randomised Phase 3 Trial. Eur Urol 2023; 83:475. [PMID: 36609010 DOI: 10.1016/j.eururo.2022.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023]
|
34
|
Kim WT, Kim J, Kim WJ. How can we best manage biochemical failure after radical prostatectomy? Investig Clin Urol 2022; 63:592-601. [PMID: 36347548 PMCID: PMC9643724 DOI: 10.4111/icu.20220294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022] Open
Abstract
Biochemical recurrence (BCR) is common after radical prostatectomy, but effective treatment options for men with BCR after curative treatment remain controversial. Although prostate-specific antigen is widely used as a surrogate marker for prostate cancer survival, it cannot fully differentiate between prostate-cancer-specific survival and overall survival. Thus, it is challenging for physicians to determine the timing of treatment to halt or slow the clinical progression of disease in patients with BCR while avoiding overtreatment for patients whose disease may not progress beyond BCR. Adjuvant therapy for radical prostatectomy or radiotherapy in intermediate- or high-risk localized prostate cancer has a benefit in terms of disease progression and survival but is not recommended in low-risk prostate cancer because of the significant adverse effects related to radiotherapy and androgen-deprivation therapy (ADT). Salvage radiotherapy (SRT) is also recommended for patients with BCR after radical prostatectomy. Several options for management of BCR after radical prostatectomy include SRT to the prostatic bed and/or pelvis, continuous or intermittent ADT, or observation. Patients' comorbidity, preferences, and cancer-related factors must be considered when deciding the best management strategy. Modern imaging technology such as positron emission tomography imaging of prostate-specific membrane antigen-positive regions enables earlier detection of disease progression, thus enhancing decision making for future disease management.
Collapse
Affiliation(s)
- Won Tae Kim
- Department of Urology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Jiyeon Kim
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University College of Medicine, Cheongju, Korea.,Institute of Urotech, Cheongju, Korea
| |
Collapse
|
35
|
French AFU Cancer Committee Guidelines - Update 2022-2024: prostate cancer - Diagnosis and management of localised disease. Prog Urol 2022; 32:1275-1372. [DOI: 10.1016/j.purol.2022.07.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022]
|
36
|
Parker CC, Clarke NW, Catton C, Kynaston H, Cook A, Cross W, Davidson C, Goldstein C, Logue J, Maniatis C, Petersen PM, Neville P, Payne H, Persad R, Pugh C, Stirling A, Saad F, Parulekar WR, Parmar MKB, Sydes MR. RADICALS-HD: Reflections before the Results are Known. Clin Oncol (R Coll Radiol) 2022; 34:593-597. [PMID: 35810050 DOI: 10.1016/j.clon.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/01/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022]
Affiliation(s)
- C C Parker
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK.
| | - N W Clarke
- Genito-Urinary Cancer Research Group, Department of Surgery, The Christie Hospital, Manchester, UK; Department of Urology, Salford Royal Hospitals, Manchester, UK
| | - C Catton
- Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - H Kynaston
- Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - A Cook
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - W Cross
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - C Davidson
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada
| | - C Goldstein
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - J Logue
- Oncology, The Christie Hospital, Manchester, UK
| | - C Maniatis
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - P M Petersen
- Department of Oncology, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - P Neville
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - H Payne
- Oncology, University College London Hospitals, London, UK
| | - R Persad
- Bristol Urological Institute, North Bristol Hospitals, Bristol, UK
| | - C Pugh
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - A Stirling
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - F Saad
- University of Montreal Hospital Center (CHUM), Montréal, Canada
| | - W R Parulekar
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada
| | - M K B Parmar
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - M R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| |
Collapse
|
37
|
PROshot: Prone Positioning, Radiation First in Total Neoadjuvant Therapy, Palliative Craniospinal Irradiation, Long-Term Androgen Deprivation Therapy, and Combining Short-Term Androgen Deprivation Therapy and Pelvic Lymph Node Coverage With Salvage Prostate Radiation. Pract Radiat Oncol 2022; 12:387-391. [PMID: 36058617 DOI: 10.1016/j.prro.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 11/21/2022]
|
38
|
Sutera P, Deek MP, Van der Eecken K, Wyatt AW, Kishan AU, Molitoris JK, Ferris MJ, Minhaj Siddiqui M, Rana Z, Mishra MV, Kwok Y, Davicioni E, Spratt DE, Ost P, Feng FY, Tran PT. Genomic biomarkers to guide precision radiotherapy in prostate cancer. Prostate 2022; 82 Suppl 1:S73-S85. [PMID: 35657158 PMCID: PMC9202472 DOI: 10.1002/pros.24373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/30/2022] [Accepted: 04/29/2022] [Indexed: 11/08/2022]
Abstract
Our ability to prognosticate the clinical course of patients with cancer has historically been limited to clinical, histopathological, and radiographic features. It has long been clear however, that these data alone do not adequately capture the heterogeneity and breadth of disease trajectories experienced by patients. The advent of efficient genomic sequencing has led to a revolution in cancer care as we try to understand and personalize treatment specific to patient clinico-genomic phenotypes. Within prostate cancer, emerging evidence suggests that tumor genomics (e.g., DNA, RNA, and epigenetics) can be utilized to inform clinical decision making. In addition to providing discriminatory information about prognosis, it is likely tumor genomics also hold a key in predicting response to oncologic therapies which could be used to further tailor treatment recommendations. Herein we review select literature surrounding the use of tumor genomics within the management of prostate cancer, specifically leaning toward analytically validated and clinically tested genomic biomarkers utilized in radiotherapy and/or adjunctive therapies given with radiotherapy.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew P. Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Kim Van der Eecken
- Department of Pathology, Ghent University Hospital, Cancer Research Institute (CRIG), Ghent, Belgium
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amar U. Kishan
- Department of Radiation Oncology, UCLA, Los Angeles, CA, USA
| | - Jason K. Molitoris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew J. Ferris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M. Minhaj Siddiqui
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zaker Rana
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark V. Mishra
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Young Kwok
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Daniel E. Spratt
- Department of Radiation Oncology, University Hospitals, Cleveland, OH, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, Antwerp, Belgium and Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Felix Y. Feng
- Departments of Radiation Oncology, Medicine and Urology, UCSF, San Francisco, CA, USA
| | - Phuoc T. Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Bratt O, Carlsson S, Fransson P, Kindblom J, Stranne J, Karlsson CT. The Swedish national guidelines on prostate cancer, part 2: recurrent, metastatic and castration resistant disease. Scand J Urol 2022; 56:278-284. [PMID: 35798533 DOI: 10.1080/21681805.2022.2093396] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVE There is now an unprecedented amount of evidence to consider when revising prostate cancer guidelines. We believe that there is a value in publishing summaries of national clinical guidelines in English for others to read and comment on. METHODS This is part 2 of a summary of the Swedish prostate cancer guidelines that were published in June 2022. This part covers recurrence after local treatment and management of metastatic and castration resistant disease. Part 1 covers early detection, diagnostics, staging, patient support and management of non-metastatic disease. RESULTS The 2022 Swedish guidelines include several new recommendations. Among these is a recommendation of a period of observation with repeated PSA tests for patients with approximately 10 years' life expectancy who experience a BCR more than 2-5 years after radical prostatectomy, to allow for estimating the PSA doubling time before deciding whether to give salvage radiotherapy or not. Recent results from the PEACE-1 trial led to the recommendation of triple-treatment with a GnRH agonist, abiraterone plus prednisolone and 6 cycles of docetaxel for patients with high-volume metastatic disease who are fit for chemotherapy. The Swedish guidelines differ from the European ones by having more restrictive recommendations about genetic testing of and high-dose zoledronic acid or denosumab treatment for men with metastatic prostate cancer, and by recommending considering bicalutamide monotherapy for selected patients with low-volume metastatic disease. CONCLUSIONS The 2022 Swedish prostate cancer guidelines include several new recommendations and some that differ from the European guidelines.
Collapse
Affiliation(s)
- Ola Bratt
- Department of Urology, Sahlgrenska University Hospital, Göteborg, Sweden.,Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Göteborg, Sweden
| | - Stefan Carlsson
- Section of Urology, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Per Fransson
- Department of Nursing, Umeå University, Umeå, Sweden
| | - Jon Kindblom
- Department of Oncology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Johan Stranne
- Department of Urology, Sahlgrenska University Hospital, Göteborg, Sweden.,Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Göteborg, Sweden
| | | | | |
Collapse
|
40
|
Tree A, Griffin C, Syndikus I, Birtle A, Choudhury A, Graham J, Ferguson C, Khoo V, Malik Z, O'Sullivan J, Panades M, Parker C, Rimmer Y, Scrase C, Staffurth J, Dearnaley D, Hall E. Nonrandomized Comparison of Efficacy and Side Effects of Bicalutamide Compared With Luteinizing Hormone-Releasing Hormone (LHRH) Analogs in Combination With Radiation Therapy in the CHHiP Trial. Int J Radiat Oncol Biol Phys 2022; 113:305-315. [PMID: 35017008 PMCID: PMC9119688 DOI: 10.1016/j.ijrobp.2021.12.160] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/19/2021] [Accepted: 12/23/2021] [Indexed: 12/02/2022]
Abstract
PURPOSE CHHiP is a randomized trial evaluating moderately hypofractionated radiation therapy for treatment of localized prostate cancer. Of all participants, 97% of them had concurrent short-course hormone therapy (HT), either luteinizing hormone-releasing hormone analog (LHRHa) or 150 mg of bicalutamide daily. This exploratory analysis compares efficacy and side effects in a nonrandomized comparison. METHODS AND MATERIALS In our study, 2700 patients received LHRHa and 403 received bicalutamide. The primary endpoint was biochemical/clinical failure. Groups were compared with Cox regression adjusted for various prognostic factors and stratified by radiation therapy dose. A key secondary endpoint was erectile dysfunction (ED) assessed by clinicians (using scores from Late Effects on Normal Tissues: Subjective/Objective/Management [LENT-SOM] subjective erectile function for vaginal penetration) and patients (single items within the University of California-Los Angeles Prostate Cancer Index [UCLA PCI] and Expanded Prostate Cancer Index Composite [EPIC]-50 questionnaires) at 2 years and compared between HT regimens by χ2 trend test. RESULTS Bicalutamide patients were significantly younger (median 67 vs 69 years LHRHa). Median follow-up was 9.3 years. There was no difference in biochemical or clinical failure with an adjusted hazard ratio or 0.97 (95% confidence interval, 0.77-1.23; P = .8). At 2 years, grade ≥2 LENT-SOM ED was reported in significantly more LHRHa patients (313 out of 590; 53%) versus bicalutamide (17 out of 68; 25%) (P < .0001). There were no differences in ED seen with UCLA-PCI and EPIC-50 questionnaires. CONCLUSIONS In this nonrandomized comparison, there was no evidence of a difference in efficacy according to type of HT received. Bicalutamide preserved clinician assessed (LENT-SOM) erectile function at 2 years but patient-reported outcomes were similar between groups.
Collapse
Affiliation(s)
- Alison Tree
- Royal Marsden NHS Foundation Trust, London, United Kingdom; Institute of Cancer Research, London, United Kingdom.
| | - Clare Griffin
- Institute of Cancer Research, London, United Kingdom
| | | | | | | | - John Graham
- Beatson Oncology Centre, Glasgow, United Kingdom
| | | | - Vincent Khoo
- Royal Marsden NHS Foundation Trust, London, United Kingdom; Institute of Cancer Research, London, United Kingdom
| | - Zafar Malik
- Whiston Hospital, Merseyside, United Kingdom
| | - Joe O'Sullivan
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | | | - Chris Parker
- Royal Marsden NHS Foundation Trust, London, United Kingdom; Institute of Cancer Research, London, United Kingdom
| | | | | | - John Staffurth
- Cardiff University/Velindre Cancer Centre, Cardiff, United Kingdom
| | - David Dearnaley
- Royal Marsden NHS Foundation Trust, London, United Kingdom; Institute of Cancer Research, London, United Kingdom
| | - Emma Hall
- Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
41
|
Jia AY, Spratt DE. Bicalutamide Monotherapy With Radiation Therapy for Localized Prostate Cancer: A Non-Evidence-Based Alternative. Int J Radiat Oncol Biol Phys 2022; 113:316-319. [PMID: 35569476 DOI: 10.1016/j.ijrobp.2022.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Angela Y Jia
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
42
|
Sood A, Keeley J, Palma-Zamora I, Chien M, Corsi N, Jeong W, Rogers CG, Trinh QD, Peabody JO, Menon M, Abdollah F. Anti-Androgen Therapy Overcomes the Time Delay in Initiation of Salvage Radiation Therapy and Rescues the Oncological Outcomes in Men with Recurrent Prostate Cancer After Radical Prostatectomy: A Post Hoc Analysis of the RTOG-9601 Trial Data. Ann Surg Oncol 2022; 29:7206-7215. [PMID: 35608801 PMCID: PMC9128637 DOI: 10.1245/s10434-022-11892-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/25/2022] [Indexed: 12/19/2022]
Abstract
Background It is unknown whether the addition of anti-androgen therapy (AAT) to late salvage radiation therapy (sRT) can lead to oncological outcomes equivalent to that of early sRT in men with recurrent prostate cancer (CaP) after surgery. Methods Data on 670 men who participated in the Radiation Therapy Oncology Group (RTOG)-9601 trial and who experienced biochemical recurrence were extracted using the National Clinical Trials Network (NCTN) data archive platform. Patients were stratified into four treatment groups: early sRT (pre-sRT prostate-specific antigen [PSA] < 0.7 ng/mL) and late sRT (pre-sRT PSA ≥ 0.7 ng/mL) with/without concomitant AAT, based on cut-offs reported in the original trial. Time-varying Cox proportional hazards and Fine–Gray competing-risk regression analyses assessed the adjusted hazards of overall mortality, CaP-specific mortality, and metastasis among the four treatment groups. Results At 15-years (median follow-up of 14.7 years), for patients treated with early sRT, early sRT with AAT, late sRT, and late sRT with AAT, the overall mortality, CaP-specific mortality, and metastasis rates were 22.9, 22.8, 40.1, and 22.9% (log-rank p = 0.0039), 12.1, 3.9, 22.7, and 8.0% (Gray’s p = 0.0004), and 18.8, 14.6, 35.9, and 19.5% (Gray’s p = 0.0004), respectively. Time-varying multivariable adjusted analysis demonstrated increased hazards of overall mortality in patients receiving delayed sRT versus early sRT (hazards ratio [HR] 1.49, 95% confidence interval [CI] 1.02–2.17); however, no difference remained after the addition of concomitant AAT to late sRT (HR 0.85, 95% CI 0.55–1.32, referent early sRT). Likewise, the hazards of cancer-specific mortality and metastatic progression were worse for late sRT when compared with early sRT, but were no different after the addition of AAT to late sRT. Conclusions Poorer outcomes associated with late sRT in men with recurrent CaP may be rescued by delivery of concomitant AAT.
Collapse
Affiliation(s)
- Akshay Sood
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA. .,Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA. .,Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jacob Keeley
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA
| | | | - Michael Chien
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Nicholas Corsi
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Wooju Jeong
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Craig G Rogers
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA.,Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Quoc-Dien Trinh
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James O Peabody
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA.,Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Mani Menon
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA.,Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA.,Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Firas Abdollah
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA.,Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
43
|
Pollack A, Karrison TG, Balogh AG, Gomella LG, Low DA, Bruner DW, Wefel JS, Martin AG, Michalski JM, Angyalfi SJ, Lukka H, Faria SL, Rodrigues GB, Beauchemin MC, Lee RJ, Seaward SA, Allen AM, Monitto DC, Seiferheld W, Sartor O, Feng F, Sandler HM. The addition of androgen deprivation therapy and pelvic lymph node treatment to prostate bed salvage radiotherapy (NRG Oncology/RTOG 0534 SPPORT): an international, multicentre, randomised phase 3 trial. Lancet 2022; 399:1886-1901. [PMID: 35569466 PMCID: PMC9819649 DOI: 10.1016/s0140-6736(21)01790-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/02/2021] [Accepted: 07/29/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND In men with a detectable prostate-specific antigen (PSA) level after prostatectomy for prostate cancer, salvage prostate bed radiotherapy (PBRT) results in about 70% of patients being free of progression at 5 years. A three-group randomised trial was designed to determine whether incremental gains in patient outcomes can be achieved by adding either 4-6 months of short-term androgen deprivation therapy (ADT) to PBRT, or both short-term ADT and pelvic lymph node radiotherapy (PLNRT) to PBRT. METHODS The international, multicentre, randomised, controlled SPPORT trial was done at 283 radiation oncology cancer treatment centres in the USA, Canada, and Israel. Eligible patients (aged ≥18 years) were those who after prostatectomy for adenocarcinoma of the prostate had a persistently detectable or an initially undetectable and rising PSA of between 0·1 and 2·0 ng/mL. Patients with and without lymphadenectomy (N0/Nx) were eligible if there was no clinical or pathological evidence of lymph node involvement. Other eligibility criteria included pT2 or pT3 disease, prostatectomy Gleason score of 9 or less, and a Zubrod performance status of 0-1. Eligible patients were randomly assigned to receive PBRT alone at a dose of 64·8-70·2 Gy at 1·8 Gy per fraction daily (group 1), PBRT plus short-term ADT (group 2), or PLNRT (45 Gy at 1·8 Gy per fraction, and then a volume reduction made to the planning target volume for the remaining 19·8-25 ·2 Gy) plus PBRT plus short-term ADT (group 3). The primary endpoint was freedom from progression, in which progression was defined as biochemical failure according to the Phoenix definition (PSA ≥2 ng/mL over the nadir PSA), clinical failure (local, regional, or distant), or death from any cause. A planned interim analysis of 1191 patents with minimum potential follow-up time of 5 years applied a Haybittle-Peto boundary of p<0·001 (one sided) for comparison of 5-year freedom from progression rates between the treatment groups. This trial is registered with ClinicalTrials.gov, NCT00567580. The primary objectives of the trial have been completed, although long-term follow-up is continuing. FINDINGS Between March 31, 2008, and March 30, 2015, 1792 eligible patients were enrolled and randomly assigned to the three treatment groups (592 to group 1 [PBRT alone], 602 to group 2 [PBRT plus short-term ADT], and 598 to group 3 [PLNRT plus PBRT plus short-term ADT]). 76 patients subsequently found to be ineligible were excluded from the analyses; thus, the evaluable patient population comprised 1716 patients. At the interim analysis (n=1191 patients; data cutoff May 23, 2018), the Haybittle-Peto boundary for 5-year freedom from progression was exceeded when group 1 was compared with group 3 (difference 17·9%, SE 2·9%; p<0·0001). The difference between groups 2 and 3 did not exceed the boundary (p=0·0063). With additional follow-up beyond the interim analysis (the final planned analysis; data cutoff May 26, 2021), at a median follow-up among survivors of 8·2 years (IQR 6·6-9·4), the 5-year freedom from progression rates in all 1716 eligible patients were 70·9% (95% CI 67·0-74·9) in group 1, 81·3% (78·0-84·6) in group 2, and 87·4% (84·7-90·2) in group 3. Per protocol criteria, freedom from progression in group 3 was superior to groups 1 and 2. Acute (≤3 months after radiotherapy) grade 2 or worse adverse events were significantly more common in group 3 (246 [44%] of 563 patients) than in group 2 (201 [36%] of 563; p=0·0034), which, in turn, were more common than in group 1 (98 [18%] of 547; p<0·0001). Similar findings were observed for grade 3 or worse adverse events. However, late toxicity (>3 months after radiotherapy) did not differ significantly between the groups, apart from more late grade 2 or worse blood or bone marrow events in group 3 versus group 2 (one-sided p=0·0060) attributable to the addition of PLNRT in this group. INTERPRETATION The results of this randomised trial establish the benefit of adding short-term ADT to PBRT to prevent progression in prostate cancer. To our knowledge, these are the first such findings to show that extending salvage radiotherapy to treat the pelvic lymph nodes when combined with short-term ADT results in meaningful reductions in progression after prostatectomy in patients with prostate cancer. FUNDING National Cancer Institute.
Collapse
Affiliation(s)
- Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL, USA.
| | - Theodore G Karrison
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA; NRG Oncology, Philadelphia, PA, USA
| | | | - Leonard G Gomella
- Sidney Kimmel Cancer Center of Thomas Jefferson University, Philadelphia, PA, USA
| | - Daniel A Low
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Deborah W Bruner
- Nell Hodgson Woodruff School of Nursing, and Winship Cancer Institute at Emory University, Atlanta, GA, USA
| | - Jeffrey S Wefel
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andre-Guy Martin
- CHU de Quebec-Université Laval (L'Hotel-Dieu de Quebec), Quebec, QC, Canada
| | - Jeff M Michalski
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Steve J Angyalfi
- Tom Baker Cancer Center, University of Calgary, Calgary, AB, Canada
| | - Himanshu Lukka
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | | | - George B Rodrigues
- Department of Oncology, London Regional Cancer Program, Western University, London, ON, Canada
| | - Marie-Claude Beauchemin
- Department of Radiation Oncology, CHUM-Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - R Jeffrey Lee
- Intermountain Medical Center, Salt Lake City, UT, USA
| | | | - Aaron M Allen
- Davidoff Center, Rabin Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Drew C Monitto
- Spartanburg Regional Medical Center, Spartanburg, SC, USA
| | | | - Oliver Sartor
- Department of Medicine, Tulane University, New Orleans, LA, USA
| | - Felix Feng
- Department of Radiation Oncology, University of California at San Francisco, San Francisco, CA , USA
| | | |
Collapse
|
44
|
Simon NI, Parker C, Hope TA, Paller CJ. Best Approaches and Updates for Prostate Cancer Biochemical Recurrence. Am Soc Clin Oncol Educ Book 2022; 42:1-8. [PMID: 35503984 PMCID: PMC9844546 DOI: 10.1200/edbk_351033] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Biochemical recurrence develops in almost one-third of men with prostate cancer after treatment with local therapy. There are numerous options for management, including surveillance, salvage radiation, androgen deprivation therapy (ADT), and clinical trials. This article reviews the current approaches to radiation therapy, ADT, and molecular imaging in men with biochemically recurrent prostate cancer. First, radiation therapy, including selection of field, dose, and use of concurrent antiandrogen therapy, is reviewed. Next, molecular imaging is addressed, including prostate-specific membrane antigen PET imaging and its increased sensitivity in identifying sites of disease. Finally, the factors associated with starting ADT are explored, and the data supporting intermittent over continuous ADT are reviewed. Lastly, the use of prostate-specific membrane antigen PET imaging and its potential role influencing therapy are discussed.
Collapse
Affiliation(s)
- Nicholas I Simon
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Chris Parker
- Royal Marsden NHS Foundation Trust, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
| | - Thomas A Hope
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA
- Department of Radiology, San Francisco VA Medical Center, San Francisco, CA
| | - Channing J Paller
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
45
|
Asso R, Degrande F, Fernandes da Silva J, Leite E. Postoperative radiotherapy in prostate cancer: When and how? – An update review. Cancer Radiother 2022; 26:742-748. [DOI: 10.1016/j.canrad.2021.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/15/2021] [Accepted: 10/12/2021] [Indexed: 11/17/2022]
|
46
|
Terlizzi M, Limkin EJ, Moukasse Y, Blanchard P. Adjuvant or Salvage Radiation Therapy for Prostate Cancer after Prostatectomy: Current Status, Controversies and Perspectives. Cancers (Basel) 2022; 14:cancers14071688. [PMID: 35406460 PMCID: PMC8996903 DOI: 10.3390/cancers14071688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary The management of patients with biochemical recurrence after prostatectomy has undergone significant changes in recent years. Currently, close monitoring of prostate-specific antigen (PSA) with early salvage radiotherapy (RT) in case of recurrence is the standard of care based on several randomized trials and a meta-analysis that has demonstrated its non-inferiority to adjuvant RT. Uncertainties remain regarding the management of patients at very high risk of recurrence, including appropriate selection criteria for adjuvant hormone therapy, and the role of imaging in refining the treatment strategy. This review explains this paradigm shift, raises points of controversy, and suggests ways to think about the future. Abstract Nearly one-third of the patients who undergo prostatectomy for prostate cancer have a biochemical recurrence (BCR) during follow-up. While several randomized trials have shown that adjuvant radiation therapy (aRT) improves biochemical control, this strategy has not been widely used because of the risk of toxicity and the fear of overtreating patients who would not have relapsed. In addition, the possibility of close PSA monitoring in the era of ultrasensitive assays enables to anticipate early salvage strategies (sRT). Three recent randomized trials and their meta-analysis have confirmed that aRT does not improve event-free survival compared to sRT, imposing the latter as the new standard of treatment. The addition of androgen deprivation therapy (ADT) to RT has been shown to improve biochemical control and metastasis-free survival, but the precise definition of to whom it should be proposed is still a matter of debate. The development of genomic tests or the use of artificial intelligence will allow more individualized treatment in the future. Therapeutic intensification with the combination of new-generation hormone therapy and RT is under study. Finally, the growing importance of metabolic imaging (PET/CT) due to its performance especially for low PSA levels will help in further personalizing management strategies.
Collapse
|
47
|
Arruda Viani G, Guimaraes Gouveia A, Trigo Leite E, Ynoe Moraes F. Moderate hypofractionation for salvage radiotherapy (HYPO-SRT) in patients with biochemical recurrence after prostatectomy: a cohort study with meta-analysis. Radiother Oncol 2022; 171:7-13. [DOI: 10.1016/j.radonc.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 01/23/2023]
|
48
|
Narrative Review of the Post-Operative Management of Prostate Cancer Patients: Is It Really the End of Adjuvant Radiotherapy? Cancers (Basel) 2022; 14:cancers14030719. [PMID: 35158986 PMCID: PMC8833528 DOI: 10.3390/cancers14030719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Among patients with prostate cancer who have been operated on, a subset harboring high-risk features will present with a biochemical recurrence (BCR). Adjuvant radiotherapy (aRT) was proven to significantly reduce the risk of BCR when compared to salvage radiotherapy (SRT) but suffered from several limitations: a lack of patient selection criteria, a higher treatment-related morbidity and an uncertain benefit for long-term clinical endpoints. In the same clinical setting, early SRT (eSRT) appears as non-inferior to aRT with a lower morbidity, replacing aRT as the preferred option. In this review, we insist on the need for multidisciplinary discussions to fully comprehend the individual characteristics of each patient and propose the best treatment strategy for every patient. Abstract Despite three randomized trials indicating a significant reduction in biochemical recurrence (BCR) in high-risk patients, adjuvant radiotherapy (aRT) was rarely performed, even in patients harboring high-risk features. aRT is associated with a higher risk of urinary incontinence and is often criticized for the lack of patient selection criteria. With a BCR rate reaching 30–70% in high-risk patients, a consensus between urologists and radiation oncologists was needed, leading to three different randomized trials challenging aRT with early salvage radiotherapy (eSRT). In these three different randomized trials with event-free survival as the primary outcome and a planned meta-analysis, eSRT appeared as non-inferior to aRT, answering, for some, this never-ending question. For many, however, the debate persists; these results raised several questions among urologists and radiation oncologists. BCR is thought to be a surrogate for clinically meaningful endpoints such as overall survival and cancer-specific survival but may be poorly efficient in comparison with metastasis-free survival. Imaging of rising prostate-specific antigen (PSA), post-operative persistent PSA and BCR was revolutionized by the broader use of MRI and nuclear imaging such as PET-PSMA; these imaging modalities were not analyzed in the previous randomized trials. A sub-group of very high-risk patients could possibly benefit from an adjuvant radiotherapy; but their usual risk factors such as high Gleason score or invaded surgical margins mean they are unable to be selected. More precise biomarkers of early BCR or even metastatic-relapse were developed in this setting and could be useful for the patients’ stratification. In this review, we insist on the need for multidisciplinary discussions to fully comprehend the individual characteristics of each patient and propose the best treatment strategy for every patient.
Collapse
|
49
|
Gonzalez-Moya A, Supiot S, Seegers V, Lizée T, Legouté F, Perennec T, Calais G. Mapping of Recurrence Sites Following Adjuvant or Salvage Radiotherapy for Prostate Cancer Patients. Front Oncol 2022; 11:787347. [PMID: 35070993 PMCID: PMC8766670 DOI: 10.3389/fonc.2021.787347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Although salvage and adjuvant radiotherapy (RT) are effective in prostate cancer (PC) patients, 30%–40% of men will have disease progression. The objective was to describe the pattern of recurrence in PC patients with biochemical failure (BF) following postoperative RT. Methods We retrospectively analyzed 935 PC patients treated from 2009 to 2019 with adjuvant or salvage RT at the Institut de Cancérologie de l’Ouest. Of these, 205 (22%) developed BF of whom 166 underwent imaging. Patients with identified radiologic failure prior any specific treatment were included to determine the site of relapse categorized as local (L)-only, locoregional (LR), or metastatic (M) recurrence. Main disease characteristics and RT fields were examined in relation to sites of recurrence. Results One hundred forty-one patients were identified with 244 sites of failure on imaging. Of these, 108 patients had received RT to the PB alone and 33 RT to the PB and pelvic lymph nodes (PB+PLN). Androgen-deprivation therapy was used concomitantly in 50 patients (35%). The median PSA at imaging was 1.6 ng/ml (range, 0–86.7). In all, 74 patients (52%) had M disease (44% in the PB group and 79% in the PB+PLN group), 61 (43%) had LR failure (52% in the PB alone group and 15% in the PB+PLN group), and six (4%) had L-only failure, at a median of 26.7 months (range, 5–110.3) from RT. Metastases were in extra-pelvic LN (37 (15%)), bones (66 (27%)), and visceral organs (eight (3%)). Fifty-three (48%) of the pelvic LN failures in the PB group would have been encompassed by standard PLN RT volume. Conclusion We found that most patients evaluated for BF after postoperative RT recurred outside the RT field. Isolated pelvic nodal failure was rare in those receiving RT to the PB+PLN but accounted for half of failures in those receiving PB alone RT. Imaging directed salvage treatment could be helpful to personalize radiation therapy plan.
Collapse
Affiliation(s)
- Ana Gonzalez-Moya
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Angers, France
| | - Stéphane Supiot
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Valérie Seegers
- Department of Medical Biostatistics, Institut de Cancérologie de l'Ouest, Angers, France
| | - Thibaut Lizée
- Department of Radiation Oncology, Jean Bernard Center, Inter-Regional Institute of Oncology, Le Mans, France
| | - Florence Legouté
- Department of Radiation Oncology, Hospital Center, La Roche sur Yon, France
| | - Tanguy Perennec
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Gilles Calais
- Department of Radiation Oncology, University Hospital Center, Tours, France
| |
Collapse
|
50
|
Management of patients with a persistently elevated PSA after radical prostatectomy: a narrative review. World J Urol 2022; 40:965-972. [PMID: 35024944 DOI: 10.1007/s00345-021-03923-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/28/2021] [Indexed: 10/19/2022] Open
Abstract
INTRODUCTION The management of the postoperative biological relapse of prostate cancer is most often based on salvage radiotherapy (SRT) with or without the addition of a variable duration of hormone therapy (HT). The indications for SRT +/- HT are established in the setting of a rising PSA level after a period where an undetectable PSA was achieved. However, in case of detectable PSA immediately after radical prostatectomy, the treatment options and prognosis are still unclear. MATERIALS AND METHODS We conducted a narrative review based on an analysis of the literature focusing on articles targeting the population of patients with postoperative persistently detectable PSA level. Case reports, original articles, clinical trials, and published reviews were studied for this purpose. CONCLUSION This article will describe current management of patients with detectable PSA immediately after radical prostatectomy, notably the contribution of modern imaging and new treatment options involving the combination of RT and HT.
Collapse
|