1
|
Poutanen M, Hagberg Thulin M, Härkönen P. Targeting sex steroid biosynthesis for breast and prostate cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00609-y. [PMID: 37684402 DOI: 10.1038/s41568-023-00609-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2023] [Indexed: 09/10/2023]
Affiliation(s)
- Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.
- Turku Center for Disease Modelling, University of Turku, Turku, Finland.
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland.
| | - Malin Hagberg Thulin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pirkko Härkönen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
2
|
Gu C, Wang Z, Lin T, Liu Z, Han W, Zhang X, Liang C, Liu H, Yu Y, Xu Z, Liu S, Wang J, Jia L, Yao X, Liao W, Fu C, Tan Z, He G, Zhu G, Fan R, Yang W, Chen X, Liu Z, Zhong L, Shi B, Ding D, Chen S, Wei J, Yao X, Chen M, Lu Z, Xie Q, Hu Z, Wang Y, Guo H, Fan T, Liang Z, Chen P, Wang W, Xu T, Li C, Xing J, Liao H, He D, Wu Z, Yu J, Feng Z, Yang M, Dou Q, Zeng Q, Li Y, Gou X, Zhou G, Wang X, Zhu R, Zhang Z, Zhang B, Tan W, Qu X, Sun H, Gan T, Ye D. Efficacy and safety of LY01005 versus goserelin implant in Chinese patients with prostate cancer: A multicenter, randomized, open-label, phase III, non-inferiority trial. Chin Med J (Engl) 2023; 136:1207-1215. [PMID: 37010251 PMCID: PMC10278759 DOI: 10.1097/cm9.0000000000002638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND LY01005 (Goserelin acetate sustained-release microsphere injection) is a modified gonadotropin-releasing hormone (GnRH) agonist injected monthly. This phase III trial study aimed to evaluated the efficacy and safety of LY01005 in Chinese patients with prostate cancer. METHODS We conducted a randomized controlled, open-label, non-inferiority trial across 49 sites in China. This study included 290 patients with prostate cancer who received either LY01005 or goserelin implants every 28 days for three injections. The primary efficacy endpoints were the percentage of patients with testosterone suppression ≤50 ng/dL at day 29 and the cumulative probability of testosterone ≤50 ng/dL from day 29 to 85. Non-inferiority was prespecified at a margin of -10%. Secondary endpoints included significant castration (≤20 ng/dL), testosterone surge within 72 h following repeated dosing, and changes in luteinizing hormone, follicle-stimulating hormone, and prostate specific antigen levels. RESULTS On day 29, in the LY01005 and goserelin implant groups, testosterone concentrations fell below medical-castration levels in 99.3% (142/143) and 100% (140/140) of patients, respectively, with a difference of -0.7% (95% confidence interval [CI], -3.9% to 2.0%) between the two groups. The cumulative probabilities of maintaining castration from days 29 to 85 were 99.3% and 97.8%, respectively, with a between-group difference of 1.5% (95% CI, -1.3% to 4.4%). Both results met the criterion for non-inferiority. Secondary endpoints were similar between groups. Both treatments were well-tolerated. LY01005 was associated with fewer injection-site reactions than the goserelin implant (0% vs . 1.4% [2/145]). CONCLUSION LY01005 is as effective as goserelin implants in reducing testosterone to castration levels, with a similar safety profile. TRIAL REGISTRATION ClinicalTrials.gov, NCT04563936.
Collapse
Affiliation(s)
- Chengyuan Gu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Zhiyu Liu
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, China
| | - Weiqing Han
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Center, Changsha, Hunan 410013, China
| | - Xuhui Zhang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, China
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hao Liu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Yang Yu
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, China
| | - Zhenzhou Xu
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Center, Changsha, Hunan 410013, China
| | - Shuang Liu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, China
| | - Jingen Wang
- Department of Urology, Jiangxi Provincial People's Hospital, School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Linghua Jia
- Department of Urology, Jiangxi Provincial People's Hospital, School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xin Yao
- Department of Urology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Wenfeng Liao
- Department of Urology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Cheng Fu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110043, China
| | - Zhaohui Tan
- Department of Urology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia 010010, China
| | - Guohua He
- Department of Urinary Surgery, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, China
| | - Guoxi Zhu
- Department of Urology, Linfen People's Hospital, Linfen, Shanxi 041000, China
| | - Rui Fan
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450006, China
| | - Wenzeng Yang
- Department of Urology, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| | - Xin Chen
- Department of Urology, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, Inner Mongolia 024000, China
| | - Zhizhong Liu
- Department of Urology, Baogang Hospital, Baotou, Inner Mongolia 014010, China
| | - Liqiang Zhong
- Department of Oncology, The Second People's Hospital of Yibin, Yibin, Sichuan 644000, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong 250014, China
| | - Degang Ding
- Department of Urology, Henan Provincial People's Hospital, The People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Shubo Chen
- Department of Urology, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei 054001, China
| | - Junli Wei
- Department of Urology, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei 054001, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhanpeng Lu
- Department of Urology, Jining No. 1 People's Hospital, Jining, Shandong 272011, China
| | - Qun Xie
- Department of Urology, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong 519000, China
| | - Zhiquan Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Tiwu Fan
- Department of Urology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046011, China
| | - Zhaozhao Liang
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Peng Chen
- Department of Urology, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Wei Wang
- Department of Urology Surgery, Liuzhou People's Hospital Affiliated to Guangxi University of Science and Technology, Liuzhou, Guangxi 545200, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
| | - Chunsheng Li
- Department of Urology, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia 024036, China
| | - Jinchun Xing
- Department of Urology Surgery, The Key Laboratory of Urinary Tract Tumors and Calculi, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361003, China
| | - Hong Liao
- Department of Urology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, Sichuan 610047, China
| | - Dalin He
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhibin Wu
- Department of Oncology, Affiliated Zhejiang Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310030, China
| | - Jiandi Yu
- Department of Urology, Zhejiang Hospital, Hangzhou, Zhejiang 310030, China
| | - Zhongwen Feng
- Department of Urology, Grand Hospital of Jincheng, Jincheng, Shanxi 048006, China
| | - Mengxiang Yang
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, Shandong 252004, China
| | - Qifeng Dou
- Department of Urology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Quan Zeng
- Department of Urology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Yuanwei Li
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Guangchen Zhou
- Department of Urology, Subei People's Hospital of Jiangsu Province (Clinical Medical College, Yangzhou University), Yangzhou, Jiangsu 225001, China
| | - Xiaofeng Wang
- Department of Urology, Peking University International Hospital, Beijing 102206, China
| | - Rujian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201300, China
| | - Zhonghua Zhang
- Department of Urology, Xinyu People's Hospital, Xinyu, Jiangxi 338000, China
| | - Bo Zhang
- Department of Urology, Tangdu Hospital, The Air Force Medical University, Xi'an, Shaanxi 710038, China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, China
| | - Xueling Qu
- Clinical Research Center of Luye Pharma Group Ltd, Beijing 264003, China
| | - Hongliang Sun
- Clinical Research Center of Luye Pharma Group Ltd, Beijing 264003, China
| | - Tianyi Gan
- Clinical Research Center of Luye Pharma Group Ltd, Beijing 264003, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
3
|
Beck J, Rouleau M, Lemire F, Neveu B, Déry M, Thériault B, Dubois G, Guérette D, Pouliot F. Mass spectrometry redefines optimal testosterone thresholds in prostate cancer patients undergoing androgen deprivation therapy. Prostate 2023; 83:670-677. [PMID: 36851864 DOI: 10.1002/pros.24501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/18/2023] [Accepted: 02/14/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) is the standard of care for prostate cancer treatment. Studies suggest that patients with testosterone levels below 0.7 nM have a longer time to castration resistance. Using the most accurate testosterone measurement method, namely mass spectrometry (MS), we sought to determine if a lower testosterone level under ADT could be associated with longer time to castration resistance. METHODS This retrospective study included 138 prostate cancer patients undergoing noncurative continuous ADT for which we had access to testosterone measurements assessed by MS. For 108 samples, paired immunoassays (IA) testosterone measurement was available. Primary outcome was time to castration-resistant prostate cancer (CRPC). The Contal and O'Quigley method was used to determine the optimal testosterone castration cut-off point considering the outcome and time-to-event variables. Relationship between testosterone levels assessed either by IA or MS and time to CRPC was evaluated using Cox regression. RESULTS Mean testosterone level was 0.370 nM by IA and 0.275 nM as assessed by MS. The optimal testosterone cut-off point identified to predict time to CRPC was of 0.705 nM for IA and of 0.270 nM for MS. While no significant difference for time to CRPC was found between patients showing IA testosterone level ≥0.705 nM versus <0.705 nM (hazard ratio [HR]: 1.579; 95% confidence interval [CI]: 0.908-2.745), patients with MS testosterone ≥0.270 nM had an increased risk of progression to CRPC compared to MS testosterone <0.270 nM in univariate (HR: 1.717; 95% CI: 1.160-2.541) and multivariate analysis (HR: 1.662; 95% CI: 1.043-2.648). CONCLUSIONS The higher sensitivity of MS testosterone measurement methods allows the identification of a lower castration threshold and leads to early identification of patients more likely to progress to CRPC. These patients would likely benefit from treatment intensification by androgen receptor axis-targeted therapies to delay disease progression.
Collapse
Affiliation(s)
- Jérémie Beck
- Division of Urology, Department of Surgery and Cancer Research Center, Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, Québec, Canada
| | - Mélanie Rouleau
- Division of Urology, Department of Surgery and Cancer Research Center, Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, Québec, Canada
| | - Francis Lemire
- Division of Urology, Department of Surgery and Cancer Research Center, Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, Québec, Canada
| | - Bertrand Neveu
- Division of Urology, Department of Surgery and Cancer Research Center, Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, Québec, Canada
| | - Michel Déry
- Biochemistry Service, Medical Laboratory Department, CHU de Québec-Université Laval, Québec, Québec, Canada
| | - Benoît Thériault
- Division of Urology, Department of Surgery and Cancer Research Center, Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, Québec, Canada
| | - Gabriel Dubois
- Division of Urology, Department of Surgery and Cancer Research Center, Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, Québec, Canada
| | - Dominique Guérette
- Biochemistry Service, Medical Laboratory Department, CHU de Québec-Université Laval, Québec, Québec, Canada
| | - Frédéric Pouliot
- Division of Urology, Department of Surgery and Cancer Research Center, Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, Québec, Canada
| |
Collapse
|
4
|
Lee TY, Pierrillas PB, Lin YW, de Greef R, Zandvliet AS, Schindler E, Migoya E. Population PK and Semimechanistic PK/PD Modeling and Simulation of Relugolix Effects on Testosterone Suppression in Men with Prostate Cancer. Clin Pharmacol Ther 2023; 113:124-134. [PMID: 36073238 PMCID: PMC10091729 DOI: 10.1002/cpt.2743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/21/2022] [Indexed: 12/24/2022]
Abstract
Relugolix, the first orally active, nonpeptide gonadotropin-releasing hormone receptor antagonist, is approved in the United States and the European Union for the treatment of adult patients with advanced prostate cancer. The recommended dosing regimen is a 360-mg loading dose followed by a 120-mg daily dose. Relugolix and testosterone concentration data and clinical information from two phase I studies, two phase II studies, and the phase III safety and efficacy study (HERO) were used to develop a population pharmacokinetic (PopPK) model and a semimechanistic population pharmacokinetic/pharmacodynamic (PopPK/PD) model that characterized relugolix exposure and its relationship to testosterone concentrations. Age, body weight, and Black/African American race had at most minimal effects on relugolix exposure or testosterone concentrations with no clinical relevance. Simulations using the PopPK/PD model confirmed the recommended dosing regimen of relugolix, with the median simulated testosterone concentrations predicted to achieve castration levels (< 50 ng/dL) and profound castration levels (< 20 ng/dL) by day 2 and day 9, respectively, and demonstrated that 97.3% and 85.5% of the patients remained at castration levels (< 50 ng/dL) upon temporary interruption of treatment for 7 days and 14 days, respectively. Collectively, simulations based on the PopPK and PopPK/PD models were consistent with actual data from clinical studies, reflecting the high predictiveness of the models and supporting the reliability of model-based simulations. These models can be used to provide guidance regarding dosing recommendations under various circumstances (e.g., temporary interruption of treatment, if needed) for relugolix.
Collapse
Affiliation(s)
- Tien-Yi Lee
- Myovant Sciences, Inc., Brisbane, California, USA
| | | | | | | | | | - Emilie Schindler
- Certara, Princeton, New Jersey, USA.,F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | |
Collapse
|
5
|
Sahu KK, Tripathi N, Agarwal N, Swami U. Relugolix in the management of prostate cancer. Expert Rev Anticancer Ther 2022; 22:891-902. [PMID: 35866612 DOI: 10.1080/14737140.2022.2105209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Relugolix is the first oral gonadotrophin-releasing hormone (GnRH) receptor antagonist. Based on the phase III HERO trial results, relugolix received Food and Drug Administration approval for adult patients with advanced prostate cancer (PCa). AREAS COVERED : We provide an overview of the preclinical and clinical development of relugolix and its role in the current treatment landscape of PCa. EXPERT OPINION Relugolix leads to rapid inhibition of testicular production of testosterone and its rapid recovery upon discontinuation. In the HERO trial, relugolix was associated with a superior cardiovascular safety profile compared to GnRH agonists. These attributes make relugolix a promising therapy for patients with pre-existing cardiovascular co-morbidities, those pursuing intermittent androgen deprivation therapy, and those who desire rapid testosterone recovery during "off-treatment" periods. In the HERO trial, very few patients received concomitant enzalutamide (n=17, 2.7%) or docetaxel (n<10, 1.3%). Safety of relugolix has not been established in combination with many androgen-receptor-axis targeted therapies (e.g. abiraterone, apalutamide), cabazitaxel, or lutetium Lu 177 vipivotide tetraxetan, which precludes its use in combination with these agents. In addition, being an oral drug, relugolix may also be associated with challenges of affordability, adherence, and compliance in this predominantly elderly population.
Collapse
Affiliation(s)
- Kamal Kant Sahu
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Nishita Tripathi
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Neeraj Agarwal
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Umang Swami
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
6
|
van Winden LJ, van Rossum HH. Testosterone analysis in prostate cancer patients. Adv Clin Chem 2022; 108:73-104. [PMID: 35659062 DOI: 10.1016/bs.acc.2021.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Testosterone is an essential steroid hormone associated with a wide variety of biological processes in humans. In prostate cancer, androgen signaling is an important driver of tumor cell growth. Depletion of gonadal testosterone, achieved by surgical or chemical castration, prevents androgenic signaling and temporally reduces, stops or reverses tumor growth before inevitable progression to castration-resistant prostate cancer occurs. Additional treatment strategies targeting androgenic signaling have become available, although these are without curative intent. While circulating testosterone is also associated with disease risk and potential clinical utility, the main use in the clinical lab is monitoring adequate castration and subsequent resistance to therapy. Adequate castrate testosterone concentrations are currently based on over 50 year-old double-isotope derivative assays that are disputed in automated immunoassay (IA) analysis. The debate has been further fueled with the introduction of mass spectrometry-based assays for testosterone, offering a substantial increase in sensitivity and specificity. In this review, we discuss testosterone regulation and androgen deprivation therapy in prostate cancer. We provide an overview of the developments in testosterone analysis for monitoring adequate castration and resistance to therapy. Current clinical practice and future clinical utility will be discussed. Finally, clinical and research recommendations will be presented.
Collapse
Affiliation(s)
- Lennart J van Winden
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Huub H van Rossum
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Merseburger AS, Roesch MC. Advanced delivery of leuprorelin acetate for the treatment of prostatic cancer. Expert Rev Anticancer Ther 2022; 22:703-715. [PMID: 35612551 DOI: 10.1080/14737140.2022.2082947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Androgen-deprivation therapy (ADT) is the main therapy for patients with advanced and metastatic prostate cancer (PCa) and, in combination with radiotherapy, for patients with localized high-risk PCa. Due to its favorable tolerability among different treatments available for ADT, leuprorelin acetate is well established as the leading luteinizing hormone-releasing hormone (LHRH) analog. The development of second-generation leuprorelin acetate (LA) depot formulation (Eligard®, Recordati S.p.A) allowed a consistent and controlled release of leuprorelin between injections and a more efficient reduction of testosterone levels with respect to conventional LHRH agonists. AREAS COVERED This work provides a summary of the biological and clinical rationale for using LA to manage PCa and presents the current evidence about the therapeutic activity of the LA gel depot formulation, used as an advanced leuprorelin acetate delivery method. EXPERT OPINION Results of the registration studies and post-marketing clinical trials demonstrate that the LA gel depot provides long-term efficacy in the clinical practice and a good degree of tolerability. Overall, collected data suggest that the LA gel depot can represent the ADT reference therapy in advanced PCa.
Collapse
|
8
|
Zastrow S, Mudra TN, Suttmann H. [Management of testosterone in advanced hormone-sensitive prostate cancer: still up to date?]. Aktuelle Urol 2022; 53:37-42. [PMID: 34555856 DOI: 10.1055/a-1525-7554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Androgen-deprivation therapy (ADT) is the standard therapy used for advanced or metastatic prostate cancer, either alone or in association with additional procedures and substances. The optimum value of testosterone postulated more than 40 years ago was arbitrarily set to be < 50 ng/dL or < 1.7 nmol/L and, from today's perspective, was defined by more insensitive measurement methods. Since then, more and more data has been generated, suggesting that a value of < 20 ng/dL would be prognostically relevant. Yet no guideline has been changed so far despite the call for lowering the target value. Measuring testosterone to evaluate the response to androgen suppression is not yet established in clinical routine. There are no specific recommendations in national and international guidelines. Based on the evolving evidence, the question about testosterone management during ADT is gaining importance. The current data is summarised in this paper.
Collapse
|
9
|
van Winden LJ, Lanfermeijer M, Heijboer AC, van Tellingen O, Bergman AM, van der Poel HG, Jonker N, van Rossum HH. Retrospective analysis of serum testosterone levels by LC-MS/MS in chemically castrated prostate cancer patients: Biological variation and analytical performance specifications. Clin Chim Acta 2021; 521:70-75. [PMID: 34217697 DOI: 10.1016/j.cca.2021.06.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND A sensitive liquid chromatography tandem-mass spectrometry (LC-MS/MS) method was used to monitor serum testosterone levels in castrated prostate cancer patients. We subsequently performed an observational and retrospective study to estimate the within- and between-subject biological variation of these patients. METHODS In total, 474 samples from 72 prostate cancer patients in the Netherlands receiving either chemical castration (CAS) or castration plus enzalutamide (ENZA) treatment were selected for data analysis. ANOVA was performed to estimate analytical variation (CVA) and within-patient variation (CVI). A nested ANOVA was applied to estimate between-patient variation (CVG). From these data, the reference change value (RCV) and analytical performance specifications (APS) were calculated. RESULTS Testosterone levels were significantly higher in the ENZA group (0.318 vs. 0.191 nmol/L, p < 0.005) than the CAS group. Overall, variation components were estimated at 6.1%, 24.6% and 60.3% for CVA, CVI and CVG, respectively. Both groups showed high individuality (<0.6). The RCV was 70.3% for all patients. Desirable APS were 12.3% for imprecision, 16.3% for bias and 26.4% for total error. CONCLUSION The generated APS are valuable for sensitive testosterone assays and the high individuality indicates that castrated testosterone levels can be studied as a predictive or prognostic biomarker in prostate cancer patients.
Collapse
Affiliation(s)
- Lennart J van Winden
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Mirthe Lanfermeijer
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annemieke C Heijboer
- Department of Clinical Chemistry: Laboratory for Endocrinology, Amsterdam University Medical Center, Location Amsterdam Medical Center and Location Free University of Amsterdam, Amsterdam, the Netherlands
| | - Olaf van Tellingen
- Department of Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andries M Bergman
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Niels Jonker
- Certe, Wilhelmina Ziekenhuis Assen, Assen, the Netherlands
| | - Huub H van Rossum
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Kouassi K, Odo B, Nzamba B, Touré P, Nziengui T, Mébiala P, Touré M. Prise en charge des effets secondaires de la suppression androgénique par castration dans le cancer de la prostate : expérience ivoirienne. PSYCHO-ONCOLOGIE 2021. [DOI: 10.3166/pson-2021-0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Contexte : L’hormonothérapie par castration dans le cancer de la prostate avancé expose le patient à des effets secondaires qui doivent être également pris en charge.
Objectif : Identifier les effets secondaires de la castration dans le cancer de la prostate avancé et leur prise en charge en milieu ivoirien.
Patients et méthode : êtude rétrospective à visée descriptive qui s’est déroulée dans le service de cancérologie du centre hospitalier de Treichville en Côte-d’Ivoire sur une période de deux ans.
Résultats : Soixante-quinze dossiers de patients ont pu être analysés. Tous les patients étaient noirs et la moitié avait moins de 55 ans. Quatre-vingt-trois pour cent des patients étaient métastatiques au diagnostic. La castration était chimique dans 84 % des cas parmi lesquelles 64 % ont reçu une suppression androgénique complète. La majorité des patients ont décrit des troubles de l’érection (100 %), des troubles de la libido (75 %), des bouffées de chaleur (88 %) et une anémie (100 %). L’anémie a été prise en charge chez tous les patients, tandis que les troubles de la sexualité ont rarement été traités.
Conclusion : La prise en charge des effets secondaires liés à l’hormonothérapie au long cours dans le cancer de la prostate doit être optimisée dans notre contexte où les patients sont jeunes et actifs.
Collapse
|
11
|
Prognostic value of testosterone castration levels following androgen deprivation and high-dose radiotherapy in localized prostate cancer: Results from a phase III trial. Radiother Oncol 2021; 160:115-119. [PMID: 33964325 DOI: 10.1016/j.radonc.2021.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND/OBJECTIVE The optimal prognostic value of testosterone following androgen deprivation therapy (ADT) is controversial. We studied the effect of serum testosterone levels on clinical outcome in localized prostate cancer (PCa) treated with ADT and high-dose radiotherapy (HRT). PATIENTS AND METHODS The DART01/05 trial randomized 355 men with intermediate and high-risk PCa to 4 months of ADT plus HRT (STADT, N = 178) or the same treatment followed by 24 months of ADT (LTADT, N = 177). This study included patients treated with LTADT who had at least 3 determinations of testosterone during ADT (N = 154). Patients were stratified into 3 subgroups by testosterone level: minimum <20 ng/dL; median 20-49 ng/dL; and maximum ≥50 ng/dL. Kaplan-Meyer and Cox regression analysis were used for overall survival (OS) and Fine & Gray regression model for metastasis free survival (MFS), biochemical disease-free survival (bDFS) and time to TT recovery. RESULTS There were no statistically significant differences in 10-year bDFS, MFS, or OS between the <20 ng/mL and 20-49 ng/dL subgroups. Multivariate analysis showed that a median testosterone ≥50 ng/dL was significantly associated with a decrease in bDFS (HR: 6.58, 95%CI 1.28-33.76, p = 0.03). Time to testosterone recovery after ADT did not correlate with bDFS, MFS, or OS and was not significantly associated with any of the testosterone subgroups. CONCLUSIONS Our results do not support the concept that additional serum testosterone suppression below 20 ng/dL is associated with better outcomes than 20-49 ng/dL. Time to testosterone recovery after ADT and HRT did not impact clinical failure.
Collapse
|
12
|
DeJongh J, Ahsman M, Snelder N. A population K-PD model analysis of long-term testosterone inhibition in prostate cancer patients undergoing intermittent androgen deprivation therapy. J Pharmacokinet Pharmacodyn 2021; 48:465-477. [PMID: 33538922 DOI: 10.1007/s10928-020-09736-7] [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: 06/25/2020] [Accepted: 12/21/2020] [Indexed: 11/25/2022]
Abstract
Intermittent androgen deprivation therapy with gonadotropin-releasing-hormone (GnRH) agonists can prevent or delay disease progression and development of castration resistant prostate cancer for subpopulations of prostate cancer patients. It may also reduce risk and severity of side effects associated with chemical castration in prostate cancer (PCa) patients. One of the earliest comprehensively documented clinical trials on this was reported in a Canadian patient population treated with leuprorelin preceded by a lead-in with cyproterone acetate. A systems-based mixed effect analysis of testosterone response in active and recovery phases allows inference of new information from this patient population. Efficacy of androgen deprivation therapy is presumed to depend on a treshold value for testosterone at the nadir, below which no additional beneficial effects on PSA reponse can be expected, and occurance of testosterone breakthroughs during active therapy. The present analysis results in a mixed effect model, incorporating GnRH receptor activation, testosterone turnover and feedback mechanisms, describing and predicting testosterone inhibition under intermittent androgen deprivation therapy on the individual and population level, during multiple years of therapy. Testosterone levels in these patients decline over time with an estimated first order rate constant of 0.083 year-1(T1/2 = 8.4 y), with a substantial distribution among this patient population, compared to the general population. PCa patients leaving the trial due to unmanageble PSA relapse appear to have slightly higher testosterone levels at the nadir than sustained responders. These findings are expected to contribute to an increased understanding of the role of testosterone in long term disease progression of prostate cancer.
Collapse
Affiliation(s)
- Joost DeJongh
- LAP&P Consultants BV, Archimedesweg 31, 2333 CM, Leiden, The Netherlands.
| | - Maurice Ahsman
- LAP&P Consultants BV, Archimedesweg 31, 2333 CM, Leiden, The Netherlands
| | - Nelleke Snelder
- LAP&P Consultants BV, Archimedesweg 31, 2333 CM, Leiden, The Netherlands
| |
Collapse
|
13
|
Sciarra A, Busetto GM, Salciccia S, Del Giudice F, Maggi M, Crocetto F, Ferro M, De Berardinis E, Scarpa RM, Porpiglia F, Carmignani L, Damiano R, Artibani W, Carrieri G. Does Exist a Differential Impact of Degarelix Versus LHRH Agonists on Cardiovascular Safety? Evidences From Randomized and Real-World Studies. Front Endocrinol (Lausanne) 2021; 12:695170. [PMID: 34194398 PMCID: PMC8237856 DOI: 10.3389/fendo.2021.695170] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022] Open
Abstract
The main systemic therapy for the management of hormone-sensitive prostate cancer (PC) is androgen deprivation therapy (ADT), with the use of long-acting luteinizing hormone releasing-hormone (LHRH) agonists considered the main form of ADT used in clinical practice to obtain castration in PC. The concomitant administration of antiandrogens for the first weeks could reduce the incidence of clinical effects related to the testosterone flare-up in the first injection of LHRH. On the contrary, Gonadotropin Rh (GnRH) antagonists produce a rapid decrease of testosterone levels without the initial flare-up, with degarelix commonly used in clinical practice to induce castration in PC patients. Even if no long-term data are reported in terms of survival to define a superiority of GnRH or LHRH, for oncological efficacy and PC control, data from randomized clinical trials and from real-life experiences, suggest a difference in cardiovascular risk of patients starting ADT. The age-related decline in testosterone levels may represent a factor connected to the increase of cardiovascular disease risk, however, the role of ADT in increasing CV events remains controversial. For these reasons, the aim of the paper is to synthesize the difference in cardiovascular risk between LHRH and degarelix in patients undergoing ADT. A difference in cardiovascular risk could be indeed an important parameter in the evaluation of these two forms of castration therapy. The Randomized trials analyzed in this paper sustain a possible protective role for degarelix versus LHRH agonists in reducing the rate of new CV events and interventions in the short-term period. On the contrary, real-word data are contradictory in different national experiences and are strongly conditioned by huge differences between the LHRH agonists group and the degarelix group.
Collapse
Affiliation(s)
- Alessandro Sciarra
- Department of Urology, Sapienza Rome University Policlinico Umberto I, Rome, Italy
| | - Gian Maria Busetto
- Department of Urology and Renal Transplantation, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Stefano Salciccia
- Department of Urology, Sapienza Rome University Policlinico Umberto I, Rome, Italy
| | | | - Martina Maggi
- Department of Urology, Sapienza Rome University Policlinico Umberto I, Rome, Italy
| | - Felice Crocetto
- Department of Neurosciences, Human Reproduction and Odontostomatology, University of Naples Federico II, Naples, Italy
- *Correspondence: Felice Crocetto,
| | - Matteo Ferro
- Department of Urology, IRCCS European Institute of Oncology (IEO), Milan, Italy
| | - Ettore De Berardinis
- Department of Urology, Sapienza Rome University Policlinico Umberto I, Rome, Italy
| | | | - Francesco Porpiglia
- Division of Urology, Department of Oncology, School of Medicine, University of Turin, San Luigi Hospital, Turin, Italy
| | - Luca Carmignani
- Department of Urology, San Donato Policlinic Hospital, Milan, Italy
| | - Rocco Damiano
- Department of Urology, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Walter Artibani
- Department of Urology, Abano Terme Policlinic, Abano Terme, Italy
| | - Giuseppe Carrieri
- Department of Urology and Renal Transplantation, University of Foggia, Policlinico Riuniti, Foggia, Italy
| |
Collapse
|
14
|
Recommandations françaises du Comité de cancérologie de l’AFU – actualisation 2020–2022 : cancer de la prostate. Prog Urol 2020; 30:S136-S251. [DOI: 10.1016/s1166-7087(20)30752-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Cornford P, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, Fanti S, Fossati N, Gandaglia G, Gillessen S, Grivas N, Grummet J, Henry AM, der Kwast THV, Lam TB, Lardas M, Liew M, Mason MD, Moris L, Oprea-Lager DE, der Poel HGV, Rouvière O, Schoots IG, Tilki D, Wiegel T, Willemse PPM, Mottet N. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer. Part II-2020 Update: Treatment of Relapsing and Metastatic Prostate Cancer. Eur Urol 2020; 79:263-282. [PMID: 33039206 DOI: 10.1016/j.eururo.2020.09.046] [Citation(s) in RCA: 662] [Impact Index Per Article: 132.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 09/24/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To present a summary of the 2020 version of the European Association of Urology (EAU)-European Association of Nuclear Medicine (EANM)-European Society for Radiotherapy & Oncology (ESTRO)-European Society of Urogenital Radiology (ESUR)-International Society of Geriatric Oncology (SIOG) guidelines on the treatment of relapsing, metastatic, and castration-resistant prostate cancer (CRPC). EVIDENCE ACQUISITION The working panel performed a literature review of the new data (2016-2019). The guidelines were updated, and the levels of evidence and/or grades of recommendation were added based on a systematic review of the literature. EVIDENCE SYNTHESIS Prostate-specific membrane antigen positron emission tomography computed tomography scanning has developed an increasingly important role in men with biochemical recurrence after local therapy. Early salvage radiotherapy after radical prostatectomy appears as effective as adjuvant radiotherapy and, in a subset of patients, should be combined with androgen deprivation. New treatments have become available for men with metastatic hormone-sensitive prostate cancer (PCa), nonmetastatic CRPC, and metastatic CRPC, along with a role for local radiotherapy in men with low-volume metastatic hormone-sensitive PCa. Also included is information on quality of life outcomes in men with PCa. CONCLUSIONS The knowledge in the field of advanced and metastatic PCa and CRPC is changing rapidly. The 2020 EAU-EANM-ESTRO-ESUR-SIOG guidelines on PCa summarise the most recent findings and advice for use in clinical practice. These PCa guidelines are first endorsed by the EANM and reflect the multidisciplinary nature of PCa management. A full version is available from the EAU office or online (http://uroweb.org/guideline/prostate-cancer/). PATIENT SUMMARY This article summarises the guidelines for the treatment of relapsing, metastatic, and castration-resistant prostate cancer. These guidelines are evidence based and guide the clinician in the discussion with the patient on the treatment decisions to be taken. These guidelines are updated every year; this summary spans the 2017-2020 period of new evidence.
Collapse
Affiliation(s)
- Philip Cornford
- Department of Urology, Liverpool University Hospitals NHS Trust, Liverpool, UK.
| | | | | | | | | | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Italy
| | - Nicola Fossati
- Unit of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giorgio Gandaglia
- Unit of Urology, Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland; University of Bern, Bern, Switzerland; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Nikolaos Grivas
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeremy Grummet
- Department of Surgery, Central Clinical School, Monash University, Caulfield North, Victoria, Australia
| | - Ann M Henry
- Leeds Cancer Centre, St. James's University Hospital and University of Leeds, Leeds, UK
| | | | - Thomas B Lam
- Academic Urology Unit, University of Aberdeen, Aberdeen, UK; Department of Urology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Michael Lardas
- Department of Urology, Metropolitan General Hospital, Athens, Greece
| | - Matthew Liew
- Department of Urology, Wrightington, Wigan and Leigh NHS Foundation Trust, Wigan, UK
| | - Malcolm D Mason
- Division of Cancer & Genetics, School of Medicine Cardiff University, Velindre Cancer Centre, Cardiff, UK
| | - Lisa Moris
- Department of Urology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Olivier Rouvière
- Hospices Civils de Lyon, Department of Urinary and Vascular Imaging, Hôpital Edouard Herriot, Lyon, France; Faculté de Médecine Lyon Est, Université de Lyon, Université Lyon 1, Lyon, France
| | - Ivo G Schoots
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Peter-Paul M Willemse
- Department of Urology, Cancer Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nicolas Mottet
- Department of Urology, University Hospital, St. Etienne, France
| |
Collapse
|
16
|
Kirby MG, Allchorne P, Appanna T, Davey P, Gledhill R, Green JSA, Greene D, Rosario DJ. Prescription switching: Rationales and risks. Int J Clin Pract 2020; 74:e13429. [PMID: 31573733 DOI: 10.1111/ijcp.13429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 09/21/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Therapeutic drug switching is commonplace across a broad range of indications and, within a drug class, is often facilitated by the availability of multiple drugs considered equivalent. Such treatment changes are often considered to improve outcomes via better efficacy or fewer side effects, or to be more cost-effective. Drug switching can be both appropriate and beneficial for several reasons; however, switching can also be associated with negative consequences. AIM To consider the impact of switching in two situations: the use of statins as a well-studied example of within-class drug switching, and gonadotropin-releasing hormone (GnRH)-targeting drug switching as an example of cross-class switching. RESULTS With the example of statins, within-class switching may be justified to reduce side effects, although the decision to switch is often also driven by the lower cost of generic formulations. With the example of GnRH agonists/antagonists, switching often occurs without the realisation that these drugs belong to different classes, with potential clinical implications. CONCLUSION Lessons emerging from these examples will help inform healthcare practitioners who may be considering switching drug prescriptions.
Collapse
Affiliation(s)
| | - Paula Allchorne
- Barts Health NHS Trust, The Royal London Hospital, London, UK
| | | | | | | | - James S A Green
- Barts Health NHS Trust, The Royal London Hospital, London, UK
| | | | | |
Collapse
|
17
|
Brassart C, Basson L, Olivier J, Latorzeff I, De Crevoisier R, Lartigau E, Pasquier D. [The radiation oncologist, one of the actors in the patient's path after cancer. Follow up after prostate cancer]. Cancer Radiother 2019; 23:565-571. [PMID: 31447344 DOI: 10.1016/j.canrad.2019.07.148] [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: 06/23/2019] [Accepted: 07/08/2019] [Indexed: 10/26/2022]
Abstract
Prostate cancer is the most common cancer of men over 50 years old. Localized prostatic cancer treatment may be responsible of a decline of patient's quality of life. The main actors of treatment are now focused on minimizing functional consequences of treatments. The radiation oncologist has a central role in patient monitoring. The follow-up is codified by official recommendations of learned societies to enhance the post-cancer period. The main objective of this article is to review the recommendations for clinical and biological follow-up. An inventory of the functional consequences of the various treatments will be detailed, and particularly those caused by androgen deprivation therapy, with a review of precautions before implementation, adverse effects and their management, as well as monitoring recommendations. The analysis of quality of life after curative treatment and suggestions to improve monitoring will also be discussed.
Collapse
Affiliation(s)
- C Brassart
- Département universitaire de radiothérapie, Centre Oscar -Lambret, 3, rue Frédéric-Combemale, 59020 Lille
| | - L Basson
- Département universitaire de radiothérapie, Centre Oscar -Lambret, 3, rue Frédéric-Combemale, 59020 Lille
| | - J Olivier
- Service d'urologie, université de Lille, CHU de Lille, 59000 Lille, France
| | - I Latorzeff
- Service d'oncologie radiothérapie, clinique Pasteur, 1, rue de la Petite-Vitesse, 31300 Toulouse, France
| | - R De Crevoisier
- Service d'oncologie radiothérapie, centre Eugène-Marquis, avenue de la Bataille-Flandre-Dunkerque, 35700 Rennes, France
| | - E Lartigau
- Département universitaire de radiothérapie, Centre Oscar -Lambret, 3, rue Frédéric-Combemale, 59020 Lille; Centre de recherche en informatique, signal et automatique de Lille UMR CNRS 9189, université de Lille, M3, avenue Carl-Gauss, 59650 Villeneuve-d'Ascq, France
| | - D Pasquier
- Département universitaire de radiothérapie, Centre Oscar -Lambret, 3, rue Frédéric-Combemale, 59020 Lille; Centre de recherche en informatique, signal et automatique de Lille UMR CNRS 9189, université de Lille, M3, avenue Carl-Gauss, 59650 Villeneuve-d'Ascq, France.
| |
Collapse
|
18
|
Ozyigit G, Hurmuz P, Yuce D, Akyol F. Prognostic significance of castrate testosterone levels for patients with intermediate and high risk prostate cancer. World J Clin Oncol 2019; 10:283-292. [PMID: 31528544 PMCID: PMC6717705 DOI: 10.5306/wjco.v10.i8.283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/05/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Testosterone level of < 50 ng/dL has been used to define castrate level after surgery or after androgen deprivation treatment (ADT) in metastatic prostate cancer (PC).
AIM To evaluate the effect of two different castrate testosterone levels, < 50 and < 20 ng/dL, on biochemical relapse free survival (BRFS) in patients with non-metastatic intermediate and high risk PC receiving definitive radiotherapy (RT) and ADT.
METHODS Between April 1998 and February 2011; 173 patients with intermediate and high risk disease were treated. Radiotherapy was delivered by either three-dimensional-conformal technique to a total dose of 73.4 Gy at the ICRU reference point or intensity modulated radiotherapy technique to a total dose of 76 Gy. All the patients received 3 mo of neoadjuvant ADT followed by RT and additional 6 mo of ADT. ASTRO Phoenix definition was used to define biochemical relapse.
RESULTS Median follow up duration was 125 months. Ninety-six patients (56%) had castrate testosterone level < 20 ng/dL and 139 patients (80%) had castrate testosterone level < 50 ng/dL. Both values are valid at predicting BRFS. However, patients with testosterone < 20 ng/dL have significantly better BRFS compared to other groups (P = 0.003). When we compare two values, it was found that using 20 ng/dL is better than 50 ng/dL in predicting the BRFS (AUC = 0.63 vs 0.58, respectively).
CONCLUSION Castrate testosterone level of less than 20 ng/dL is associated with better BRFS and is better in predicting the BRFS. Further studies using current standard of care of high dose IMRT and longer ADT duration might support these findings.
Collapse
Affiliation(s)
- Gokhan Ozyigit
- Department of Radiation Oncology, Hacettepe University Faculty of Medicine, Ankara 06100, Turkey M.D
| | - Pervin Hurmuz
- Department of Radiation Oncology, Hacettepe University Faculty of Medicine, Ankara 06100, Turkey M.D
| | - Deniz Yuce
- Department of Preventive Oncology, Hacettepe University Faculty of Medicine, Ankara 06100, Turkey
| | - Fadil Akyol
- Department of Radiation Oncology, Hacettepe University Faculty of Medicine, Ankara 06100, Turkey M.D
| |
Collapse
|
19
|
Hurmuz P, Ozyigit G. In Regard to Bryant et al. Int J Radiat Oncol Biol Phys 2019; 104:224-225. [PMID: 30967233 DOI: 10.1016/j.ijrobp.2019.01.083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 12/31/2019] [Accepted: 01/18/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Pervin Hurmuz
- Department of Radiation Oncology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Gokhan Ozyigit
- Department of Radiation Oncology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
20
|
Bryant AK, McKay RR, Kader AK, Parsons JK, Einck JP, Kane CJ, Mundt AJ, Murphy JD, Rose BS. Subcastrate Testosterone Nadir and Clinical Outcomes in Intermediate- or High-Risk Localized Prostate Cancer. Int J Radiat Oncol Biol Phys 2018; 103:1068-1076. [PMID: 30543857 DOI: 10.1016/j.ijrobp.2018.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/17/2018] [Accepted: 12/02/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE It is unclear if additional serum testosterone suppression below the castrate threshold of 50 ng/dL improves clinical outcomes in patients with localized prostate cancer undergoing definitive therapy. METHODS AND MATERIALS We examined the association of subcastrate testosterone nadir with prostate-specific antigen (PSA) response and long-term clinical outcomes in 764 U.S. veterans with intermediate- or high-risk localized prostate cancer treated with androgen deprivation therapy and definitive radiation therapy from 2000 to 2015. Patients were categorized into testosterone nadir groups based on the minimum testosterone measurement during continuous gonadotropic-releasing hormone agonist therapy (<20 ng/dL vs 20-49 ng/dL). Outcomes included PSA response (3-month post-radiation therapy PSA and 2-year PSA nadir; multivariable linear regression) and long-term clinical outcomes (biochemical recurrence, metastasis, and prostate cancer-specific mortality; Fine-Gray competing risk regression). RESULTS A testosterone nadir of 20 to 49 ng/dL was associated with higher 3-month post-radiation therapy PSA compared to <20 ng/dL (ß = 0.16, 95% confidence interval [CI], 0.06-0.26, P = .001) and higher 2-year PSA nadir (ß = 0.12, 95% CI, 0.04-0.21, P = .005). Compared to the <20-ng/dL group, the 20 to 49-ng/dL group showed higher 10-year biochemical recurrence rates (28.1% vs 18.3%) and metastasis rates (12.9% vs 7.8%) persisting on multivariable analyses (biochemical recurrence: sub-distribution hazard ratio [SDHR], 1.62 for 20-49 ng/dL, 95% CI, 1.07-2.45, P = .02; metastasis: SDHR, 2.19, 95% CI, 1.21-3.94, P = .009). There was a trend toward inferior prostate cancer-specific mortality for the 20 to 49-ng/dL group (SDHR, 1.95, 95% CI, 0.90-4.22, P = .09). CONCLUSIONS Additional serum testosterone suppression below 50 ng/dL was associated with improved PSA responses and lower rates of biochemical recurrence and metastasis in this cohort of patients with localized prostate cancer.
Collapse
Affiliation(s)
- Alex K Bryant
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | - Rana R McKay
- Division of Hematology-Oncology, Department of Internal Medicine, University of California San Diego, San Diego, California
| | - A Karim Kader
- Department of Urology, University of California San Diego, San Diego, California
| | - J Kellogg Parsons
- Department of Urology, University of California San Diego, San Diego, California
| | - John P Einck
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | - Christopher J Kane
- Department of Urology, University of California San Diego, San Diego, California
| | - Arno J Mundt
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California; Clinical and Translational Research Institute, University of California San Diego, San Diego, California
| | - James D Murphy
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California; Clinical and Translational Research Institute, University of California San Diego, San Diego, California
| | - Brent S Rose
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California; Clinical and Translational Research Institute, University of California San Diego, San Diego, California.
| |
Collapse
|
21
|
Rozet F, Hennequin C, Beauval JB, Beuzeboc P, Cormier L, Fromont-Hankard G, Mongiat-Artus P, Ploussard G, Mathieu R, Brureau L, Ouzzane A, Azria D, Brenot-Rossi I, Cancel-Tassin G, Cussenot O, Rebillard X, Lebret T, Soulié M, Penna RR, Méjean A. RETRACTED: Recommandations françaises du Comité de Cancérologie de l’AFU – Actualisation 2018–2020 : cancer de la prostate French ccAFU guidelines – Update 2018–2020: Prostate cancer. Prog Urol 2018; 28:S79-S130. [PMID: 30392712 DOI: 10.1016/j.purol.2018.08.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/31/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy).
Cet article est retiré de la publication à la demande des auteurs car ils ont apporté des modifications significatives sur des points scientifiques après la publication de la première version des recommandations.
Le nouvel article est disponible à cette adresse: DOI:10.1016/j.purol.2019.01.007.
C’est cette nouvelle version qui doit être utilisée pour citer l’article.
This article has been retracted at the request of the authors, as it is not based on the definitive version of the text because some scientific data has been corrected since the first issue was published.
The replacement has been published at the DOI:10.1016/j.purol.2019.01.007.
That newer version of the text should be used when citing the article.
Collapse
Affiliation(s)
- F Rozet
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, institut mutualiste Montsouris, université René-Descartes, 42, boulevard Jourdan, 75674, Paris, France.
| | - C Hennequin
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service de radiothérapie, Saint-Louis Hospital, AP-HP, 75010, Paris, France
| | - J-B Beauval
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, oncologie médicale, institut universitaire du cancer Toulouse-Oncopole, CHU Rangueil, 31100, Toulouse, France
| | - P Beuzeboc
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, hôpital Foch, 92150, Suresnes, France
| | - L Cormier
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, CHU François-Mitterrand, 21000, Dijon, France
| | - G Fromont-Hankard
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; CHU de Tours, 2, boulevard Tonnellé, 37000, Tours, France
| | - P Mongiat-Artus
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, hôpital Saint-Louis, 1, avenue Claude-Vellefaux, Paris cedex 10, France
| | - G Ploussard
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, clinique La Croix du Sud-Saint-Jean Languedoc, institut universitaire du cancer, 31100, Toulouse, France
| | - R Mathieu
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, hôpital de Rennes, 2, rue Henri-le-Guilloux, 35033, Rennes cedex 9, France
| | - L Brureau
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Inserm, U1085, IRSET, 97145 Pointe-à-Pitre, Guadeloupe
| | - A Ouzzane
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, hôpital Claude-Huriez, CHRU de Lille, rue Michel-Polonovski, 59000, Lille, France
| | - D Azria
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Inserm U1194, ICM, université de Montpellier, 34298, Montpellier, France
| | - I Brenot-Rossi
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Institut Paoli-Calmettes, 232, boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - G Cancel-Tassin
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; GRC no 5 ONCOTYPE-URO, institut universitaire de cancérologie, Sorbonne université, 75020, Paris, France
| | - O Cussenot
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, hôpital Tenon, AP-HP, Sorbonne université, 75020, Paris, France
| | - X Rebillard
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, clinique mutualiste Beau-Soleil, 119, avenue de Lodève, 34070, Montpellier, France
| | - T Lebret
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, hôpital Foch, 92150, Suresnes, France
| | - M Soulié
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Centre hospitalier universitaire Rangueil, 31059, Toulouse, France
| | - R Renard Penna
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; GRC no 5 ONCOTYPE-URO, institut universitaire de cancérologie, Sorbonne université, 75020, Paris, France; Service de radiologie, hôpital Tenon, AP-HP, 75020, Paris, France
| | - A Méjean
- Comité de cancérologie de l'Association française d'urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017, Paris, France; Service d'urologie, hôpital européen Georges-Pompidou, université Paris Descartes, Assistance publique des hôpitaux de Paris (AP-HP), 75015, Paris, France
| |
Collapse
|
22
|
Rozet F, Hennequin C, Beauval JB, Beuzeboc P, Cormier L, Fromont-Hankard G, Mongiat-Artus P, Ploussard G, Mathieu R, Brureau L, Ouzzane A, Azria D, Brenot-Rossi I, Cancel-Tassin G, Cussenot O, Rebillard X, Lebret T, Soulié M, Renard Penna R, Méjean A. Recommandations françaises du Comité de Cancérologie de l’AFU – Actualisation 2018–2020 : cancer de la prostate. Prog Urol 2018; 28 Suppl 1:R81-R132. [DOI: 10.1016/j.purol.2019.01.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 01/02/2023]
|
23
|
Cabarkapa S, Perera M, Sikaris K, O’Brien JS, Bolton DM, Lawrentschuk N. Reporting and ideal testosterone levels in men undergoing androgen deprivation for prostate cancer-time for a rethink? Prostate Int 2018; 6:1-6. [PMID: 29556482 PMCID: PMC5857188 DOI: 10.1016/j.prnil.2017.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 05/01/2017] [Accepted: 05/18/2017] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND This study aims to review current laboratory reporting strategies across Australia and New Zealand with a view to propose a more useful template for reporting serum testosterone in the context of prostate cancer. MATERIALS AND METHODS Registered pathology laboratories in Australia and New Zealand were enrolled into the current study. An electronic or a phone survey was utilized to collect data from each participating laboratory. Obtained information included assay utilized, units reported, reference intervals, lowest reported value, and lowest detectable value. To identify recommendations for testosterone testing, a systematic search was performed across Web of Science (including MEDLINE), EMBASE, and Cochrane libraries. RESULTS Assessment of national pathology laboratories identified significant heterogeneity in the reporting methods. Reports typically used a "normal healthy male of 35 years of age" as a comparator but did not refer to optimal castrate levels, the lowest level that their assay was able to detect, nor did they include appended clinical guidelines relating to the prostate cancer patient cohort. CONCLUSIONS Across Australia and New Zealand, various methods for testing and reporting serum testosterone exist, while international guidelines remain vague. The fashion in which serum testosterone levels are displayed should be re-evaluated to address the relevant clinical population and reflect an agreed-upon castrate threshold in patients undergoing androgen deprivation therapy.
Collapse
Affiliation(s)
- Sonja Cabarkapa
- Department of Surgery, Austin Hospital, University of Melbourne, Melbourne, Australia
| | - Marlon Perera
- Department of Surgery, Austin Hospital, University of Melbourne, Melbourne, Australia
| | - Ken Sikaris
- Department of Pathology, University of Melbourne, Melbourne, Australia
| | - Jonathan S. O’Brien
- Department of Surgery, Austin Hospital, University of Melbourne, Melbourne, Australia
| | - Damien M. Bolton
- Department of Surgery, Austin Hospital, University of Melbourne, Melbourne, Australia
- Department of Pathology, University of Melbourne, Melbourne, Australia
| | - Nathan Lawrentschuk
- Department of Surgery, Austin Hospital, University of Melbourne, Melbourne, Australia
- Olivia Newton-John Cancer Research Institute, Austin Hospital, Melbourne, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Australia
| |
Collapse
|
24
|
Poon DMC, Chan CK, Chan TW, Cheung FY, Kwong PWK, Lee EKC, Leung AKC, Leung SYL, Ma WK, So HS, Tam PC, Ho LY. Consensus statements on the management of metastatic prostate cancer from the Hong Kong Urological Association and Hong Kong Society of Uro-Oncology. BJU Int 2018; 121:703-715. [DOI: 10.1111/bju.14091] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Darren Ming-Chun Poon
- Department of Clinical Oncology; State Key Laboratory in Oncology in South China; Sir YK Pao Centre for Cancer; Hong Kong Cancer Institute and Prince of Wales Hospital; Chinese University of Hong Kong; Hong Kong Hong Kong
| | - Chi-Kwok Chan
- Department of Surgery; Prince of Wales Hospital; Hong Kong Hong Kong
| | - Tim-Wai Chan
- Department of Clinical Oncology; Queen Elizabeth Hospital; Hong Kong Hong Kong
| | | | | | - Eric Ka-Chai Lee
- Department of Clinical Oncology; Tuen Mun Hospital; Hong Kong Hong Kong
| | | | | | - Wai-Kit Ma
- Department of Surgery; Queen Mary Hospital; Hong Kong Hong Kong
| | - Hing-Shing So
- Division of Urology; Department of Surgery; United Christian Hospital; Hong Kong Hong Kong
| | - Po-Chor Tam
- Department of Surgery; Queen Mary Hospital; Hong Kong Hong Kong
| | | |
Collapse
|
25
|
Pagliarulo V. Androgen Deprivation Therapy for Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1096:1-30. [PMID: 30324345 DOI: 10.1007/978-3-319-99286-0_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the contemporary scene, less than 5% of men with newly diagnosed prostate cancer (PC) have metastases at first presentation, compared to 20-25%, more than 20 years ago. Nonetheless, the use of androgen deprivation therapy (ADT) has increased over the years, suggesting that patients in Europe and United States may receive ADT in cases of lower disease burden, and not always according to evidence based indications. Nonetheless, PC remains the second most common cause of cancer death after lung cancer in American men. Thus, there is a need for more effective, specific and well tolerated agents which can provide a longer and good quality of life while avoiding the side effects related to disease and treatment morbidity.After mentioning the current knowledge on the endocrinology of androgens and androgen receptor, relevant to PC development, as well as the possible events occurring during PC initiation, we will compare different hormonal compounds available for the treatment of PC, both from a pharmacological standpoint, and in terms of contemporary clinical indications.
Collapse
Affiliation(s)
- Vincenzo Pagliarulo
- Department of Urology, University "Aldo Moro", Bari, Italy. .,Azienda Ospedaliero-Universitaria Policlinico, Bari, Italy.
| |
Collapse
|
26
|
Morote J, Comas I, Ferrer R, Planas J, Celma A, Regis L. Accuracy of serum luteinizing hormone and serum testosterone measurements to assess the efficacy of medical castration in prostate cancer patients. J Biomed Sci 2017; 24:81. [PMID: 29058606 PMCID: PMC5651562 DOI: 10.1186/s12929-017-0386-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/07/2017] [Indexed: 01/28/2023] Open
Abstract
Background Luteinizing hormone-releasing hormone (LH-RH) agonists are the standard for androgen deprivation therapy (ADT) in prostate cancer (PCa) patients. Current guidelines recommend serum testosterone measurement to assess the efficacy of ADT and to define castration resistance. However, serum testosterone does not reflect the exclusive effect of castration due to its extratesticular production. The aim of this study is to analyze if serum LH reflects better than serum testosterone the activity of LH-RH agonists. Methods Serum LH and serum testosterone were measured with chemiluminescent immunoassay (CLIA) in a cohort study of 1091 participants: 488 PCa patients “on LH-RH agonists”, 303 “off LH-RH agonist” in whom LH-RH agonists were withdrawn, and 350 men with PCa suspicion “no LH-RH agonist” who never received LH-RH agonists. In a validation cohort of 147 PCa patients, 124 on “LH-RH agonists” and 19 “off LH-RH agonists”, serum testosterone was also measured with liquid chromatography and tandem mass spectrometry (LC MSMS). Results The area under the curve (AUC) to distinguish patients “on versus off LH-RH agonists” was 0.997 for serum LH and 0.740 for serum testosterone, P < 0.001. The 97.5 percentile of serum LH in patients “on LH-RH agonists” was 0.97 U/L, been the most efficient threshold 1.1 U/L. The AUCs for serum LH, testosterone measured with CLIA and with LC MSMS, in the validation cohort, were respectively 1.000, 0.646 and 0.814, P < 0.001. The efficacy to distinguish patients “on versus off LH-RH agonists” was 98.6%, 78.3%, and 89.5% respectively, using 1.1 U/L as threshold for serum LH and 50 ng/dL for serum testosterone regardless the method. Conclusions Serum LH is more accurate than serum testosterone regardless the method, to distinguish patients “on versus off LH-RH agonists”. The castrate level of serum LH is 1.1 U/l. These findings suggest that assessment of LH-RH agonist efficacy and castration resistance definition should be reviewed.
Collapse
Affiliation(s)
- Juan Morote
- Department of Urology, Vall d'Hebron Hospital, Universidad Autonoma de Barcelona, 14 Po Vall d'Hebron 119-129, 00173, Barcelona, Spain.
| | - Imma Comas
- Department of Biochemistry, Vall d'Hebron Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roser Ferrer
- Department of Biochemistry, Vall d'Hebron Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jacques Planas
- Department of Urology, Vall d'Hebron Hospital, Universidad Autonoma de Barcelona, 14 Po Vall d'Hebron 119-129, 00173, Barcelona, Spain
| | - Anna Celma
- Department of Urology, Vall d'Hebron Hospital, Universidad Autonoma de Barcelona, 14 Po Vall d'Hebron 119-129, 00173, Barcelona, Spain
| | - Lucas Regis
- Department of Urology, Vall d'Hebron Hospital, Universidad Autonoma de Barcelona, 14 Po Vall d'Hebron 119-129, 00173, Barcelona, Spain
| |
Collapse
|
27
|
Sayyid RK, Sayyid AK, Klaassen Z, Fadaak K, Goldberg H, Chandrasekar T, Ahmad A, Leao R, Perlis N, Chadwick K, Hamilton RJ, Kulkarni GS, Finelli A, Zlotta AR, Fleshner NE. Testosterone Responders to Continuous Androgen Deprivation Therapy Show Considerable Variations in Testosterone Levels on Followup: Implications for Clinical Practice. J Urol 2017; 199:251-256. [PMID: 28751266 DOI: 10.1016/j.juro.2017.07.078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE We determined whether men on continuous androgen deprivation therapy who achieve testosterone less than 0.7 nmol/l demonstrate subsequent testosterone elevations during followup and whether such events predict worse oncologic outcomes. MATERIALS AND METHODS We evaluated a random, retrospective sample of 514 patients with prostate cancer treated with continuous androgen deprivation therapy in whom serum testosterone was less than 0.7 nmol/l at University Health Network between 2007 and 2016. Patients were followed from the date of the first testosterone measurement of less than 0.7 nmol/l to progression to castrate resistance, death or study period end. Study outcomes were the development of testosterone elevations greater than 0.7, greater than 1.1 and greater than 1.7 nmol/l, and progression to a castrate resistant state. Survival curves were constructed to determine the rate of testosterone elevations. Multivariate Cox regression analysis was done to assess whether elevations predicted progression to castrate resistance. RESULTS Median patient age was 74 years and median followup was 20.3 months. Within 5 years of followup 82%, 45% and 18% of patients had subsequent testosterone levels greater than 0.7, greater than 1.1 and greater than 1.7 nmol/l, respectively. In 96% to 100% of these patients levels less than 0.7 nmol/l were subsequently reestablished within 5 years. No patient baseline characteristic was associated with elevations and elevations were not a significant predictor of progression to a castrate resistant state. CONCLUSIONS Men on continuous androgen deprivation therapy in whom initial testosterone is less than 0.7 nmol/l frequently show subsequent elevations in serum testosterone. Such a development should not trigger an immediate response from physicians as these events are prognostically insignificant with regard to oncologic outcomes. Levels are eventually reestablished at less than 0.7 nmol/l.
Collapse
Affiliation(s)
- Rashid K Sayyid
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Section of Urology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Abdallah K Sayyid
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Zachary Klaassen
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Section of Urology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Kamel Fadaak
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hanan Goldberg
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Thenappan Chandrasekar
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ardalanejaz Ahmad
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ricardo Leao
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Nathan Perlis
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Karen Chadwick
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Robert J Hamilton
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Girish S Kulkarni
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Antonio Finelli
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | | - Neil E Fleshner
- Division of Urology, Department of Surgical Oncology, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
28
|
Abstract
OBJECTIVES The purpose of the guidelines national committee CCAFU was to propose updated french guidelines for localized and metastatic prostate cancer (PCa). METHODS A Medline search was achieved between 2013 and 2016, as regards diagnosis, options of treatment and follow-up of PCa, to evaluate different references with levels of evidence. RESULTS Epidemiology, classification, staging systems, diagnostic evaluation are reported. Disease management options are detailed. Recommandations are reported according to the different clinical situations. Active surveillance is a major option in low risk PCa. Radical prostatectomy remains a standard of care of localized PCa. The three-dimensional conformal radiotherapy is the technical standard. A dose of > 74Gy is recommended. Moderate hypofractionation provides short-term biochemical control comparable to conventional fractionation. In case of intermediate risk PCa, radiotherapy can be combined with short-term androgen deprivation therapy (ADT). In case of high risk disease, long-term ADT remains the standard of care. ADT is the backbone therapy of metastatic disease. In men with metastases at first presentation, upfront chemotherapy combined with ADT should be considered as a new standard. In case of metastatic castration-resistant PCa (mCRPC), new hormonal treatments and chemotherapy provide a better control of tumor progression and increase survival. CONCLUSIONS These updated french guidelines will contribute to increase the level of urological care for the diagnosis and treatment for prostate cancer. © 2016 Elsevier Masson SAS. All rights reserved.
Collapse
|
29
|
Wang Y, Dai B, Ye DW. Serum testosterone level predicts the effective time of androgen deprivation therapy in metastatic prostate cancer patients. Asian J Androl 2017; 19:178-183. [PMID: 26975487 PMCID: PMC5312215 DOI: 10.4103/1008-682x.174856] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Androgen deprivation therapy (ADT) is the standard of care for patients with metastatic prostate cancer. However, whether serum testosterone levels, using a cut-off point of 50 ng dl−1, are related to the effective time of ADT in newly diagnosed prostate cancer patients remains controversial. Moreover, recent studies have shown that some patients may benefit from the addition of upfront docetaxel chemotherapy. To date, no studies have been able to distinguish patients who will benefit from the combination of ADT and docetaxel chemotherapy. This study included 206 patients who were diagnosed with metastatic prostate cancer and showed progression to castrate-resistance prostate cancer (CRPC). Serum testosterone levels were measured prospectively after ADT for 1, 3, and 6 months. The endpoint was the time to CRPC. In univariate and multivariate analyses, testosterone levels <50 ng dl−1 were not associated with the effective time of ADT. Receiver operating characteristic and univariate analysis showed that testosterone levels of ≤25 ng dl−1 after the first month of ADT offered the best overall sensitivity and specificity for prediction of a longer time to CRPC (adjusted hazard ratio [HR], 1.46; 95% confidence interval [95% CI], 1.08–1.96; P = 0.013). Our results show that serum testosterone level of 25 ng dl−1 plays a prognostic role in prostate cancer patients receiving ADT. A testosterone value of 25 ng dl−1 after the first month of ADT can distinguish patients who benefit from ADT effectiveness for only a short time. These patients may need to receive ADT and concurrent docetaxel chemotherapy.
Collapse
Affiliation(s)
- Yue Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 20032, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 20032, China
| | - Ding-Wei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 20032, China
| |
Collapse
|
30
|
Regis L, Planas J, Carles J, Maldonado X, Comas I, Ferrer R, Morote J. Free Testosterone During Androgen Deprivation Therapy Predicts Castration-Resistant Progression Better Than Total Testosterone. Prostate 2017; 77:114-120. [PMID: 27800640 DOI: 10.1002/pros.23256] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/31/2016] [Indexed: 11/06/2022]
Abstract
BACKGROUND The optimal degree of testosterone suppression in patients with prostate cancer undergoing androgen deprivation therapy remains in question. Furthermore, serum free testosterone, which is the active form of testosterone, seems to correlate with intraprostatic testosterone. Here we compared free and total serum testosterone as predictors of survival free of castration resistance. METHODS Total testosterone (chemiluminescent assay, lower sensitivity 10 ng/dl) and free testosterone (analogue-ligand radioimmunoassay, lower sensitivity 0.05 pg/ml) were determined at 6 months of LHRH agonist treatment in a prospective cohort of 126 patients with prostate cancer. During a mean follow-up of 67 months (9-120), 75 (59.5%) events of castration-resistant progression were identified. Multivariate analysis and survival analysis according to total testosterone cutoffs of 50, 32, and 20 ng/dl, and free testosterone cutoffs of 1.7, 1.1, and 0.7 pg/ml were performed. RESULTS Metastatic spread was the most powerful predictor of castration resistance, HR: 2.09 (95%CI: 1.18-3.72), P = 0.012. Gleason score, baseline PSA and PSA at 6 months were also independents predictors, but not free and total testosterone. Stratified analysis was conducted on the basis of the status of metastatic diseases and free testosterone was found to be an independent predictor of survival free of castration resistance in the subgroup of patients without metastasis, HR: 2.12 (95%CI: 1.16-3.85), P = 0.014. The lowest threshold of free testosterone which showed significant differences was 1.7 pg/ml, P = 0.003. CONCLUSIONS Free testosterone at 6 months of LHRH agonist treatment seems to be a better surrogate than total testosterone to predict castration resistance in no metastatic prostate cancer patients. Prostate 77:114-120, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lucas Regis
- Department of Urology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jacques Planas
- Department of Urology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Carles
- Department of Medical Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Maldonado
- Department of Radiation Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Inma Comas
- Department of Biochemistry, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roser Ferrer
- Department of Biochemistry, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Morote
- Department of Urology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Morote J, Regis L, Celma A, Planas J. Measurement of serum testosterone during androgenic suppression in patients with prostate cancer: A systematic review. Actas Urol Esp 2016; 40:477-84. [PMID: 26899928 DOI: 10.1016/j.acuro.2016.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Clinical practice guidelines recommend measuring serum testosterone (ST) during androgenic suppression (AS) to assess its efficacy and define castration resistance (CR). The objectives of this systematic review were to assess the level of scientific evidence that justify checking ST levels during AS, when to perform it and for what purpose. MATERIAL AND METHODS We performed a search in PubMed with the following mesh terms: androgen suppression, testosterone, and prostate cancer. The search was narrowed to original articles published in English. RESULTS We found 8 publications that analysed the clinical impact of ST concentrations during AS. In all of the series, ST was measured using chemiluminescent assays. However, only indirect methods based on liquid or gas chromatography for its extraction and subsequent quantification using mass spectrometry are recommended, especially for measuring low levels. The endpoints were specific survival and CR-free survival. Six studies were retrospective. The series were not uniform in terms of clinical stage, types of AS and ST assessment methods. In general, low ST levels (<20ng/dL or <32ng/dL) were related to longer CR-free survival. The measurements were performed every 3 or 6 months. Four studies confirmed the beneficial effect of adding bicalutamide when detecting microelevations above 50ng/dL. CONCLUSIONS The level of scientific evidence justifying the measurement of ST during AS is low, and the methods employed for quantifying ST levels are inadequate. However, we consider it useful to check ST levels during AS, and there appears to be an association between low ST levels and better disease outcomes. In the event of microelevations above 50ng/dL, we recommend the administration of bicalutamide.
Collapse
Affiliation(s)
- J Morote
- Servicio de Urología, Hospital Vall d́Hebron, Universitat Autònoma de Barcelona, Barcelona, España
| | - L Regis
- Servicio de Urología, Hospital Vall d́Hebron, Universitat Autònoma de Barcelona, Barcelona, España.
| | - A Celma
- Servicio de Urología, Hospital Vall d́Hebron, Universitat Autònoma de Barcelona, Barcelona, España
| | - J Planas
- Servicio de Urología, Hospital Vall d́Hebron, Universitat Autònoma de Barcelona, Barcelona, España
| |
Collapse
|
32
|
Cornford P, Bellmunt J, Bolla M, Briers E, De Santis M, Gross T, Henry AM, Joniau S, Lam TB, Mason MD, van der Poel HG, van der Kwast TH, Rouvière O, Wiegel T, Mottet N. EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part II: Treatment of Relapsing, Metastatic, and Castration-Resistant Prostate Cancer. Eur Urol 2016; 71:630-642. [PMID: 27591931 DOI: 10.1016/j.eururo.2016.08.002] [Citation(s) in RCA: 1104] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/02/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To present a summary of the 2016 version of the European Association of Urology (EAU) - European Society for Radiotherapy & Oncology (ESTRO) - International Society of Geriatric Oncology (SIOG) Guidelines on the treatment of relapsing, metastatic, and castration-resistant prostate cancer (CRPC). EVIDENCE ACQUISITION The working panel performed a literature review of the new data (2013-2015). The guidelines were updated, and the levels of evidence and/or grades of recommendation were added based on a systematic review of the literature. EVIDENCE SYNTHESIS Relapse after local therapy is defined by a rising prostate-specific antigen (PSA) level >0.2ng/ml following radical prostatectomy (RP) and >2ng/ml above the nadir after radiation therapy (RT). 11C-choline positron emission tomography/computed tomography is of limited importance if PSA is <1.0ng/ml; bone scans and computed tomography can be omitted unless PSA is >10ng/ml. Multiparametric magnetic resonance imaging and biopsy are important to assess biochemical failure following RT. Therapy for PSA relapse after RP includes salvage RT at PSA levels <0.5ng/ml and salvage RP, high-intensity focused ultrasound, cryosurgical ablation or salvage brachytherapy of the prostate in radiation failures. Androgen deprivation therapy (ADT) remains the basis for treatment of men with metastatic prostate cancer (PCa). However, docetaxel combined with ADT should be considered the standard of care for men with metastases at first presentation, provided they are fit enough to receive the drug. Follow-up of ADT should include analysis of PSA, testosterone levels, and screening for cardiovascular disease and metabolic syndrome. Level 1 evidence for the treatment of metastatic CRPC (mCRPC) includes, abiraterone acetate plus prednisone (AA/P), enzalutamide, radium 223 (Ra 223), docetaxel at 75 mg/m2 every 3 wk and sipuleucel-T. Cabazitaxel, AA/P, enzalutamide, and radium are approved for second-line treatment of CRPC following docetaxel. Zoledronic acid and denosumab can be used in men with mCRPC and osseous metastases to prevent skeletal-related complications. CONCLUSIONS The knowledge in the field of advanced and metastatic PCa and CRPC is changing rapidly. The 2016 EAU-ESTRO-SIOG Guidelines on PCa summarise the most recent findings and advice for use in clinical practice. These PCa guidelines are the first endorsed by the European Society for Therapeutic Radiology and Oncology and the International Society of Geriatric Oncology and reflect the multidisciplinary nature of PCa management. A full version is available from the EAU office or online (http://uroweb.org/guideline/prostate-cancer/). PATIENT SUMMARY In men with a rise in their PSA levels after prior local treatment for prostate cancer only, it is important to balance overtreatment against further progression of the disease since survival and quality of life may never be affected in many of these patients. For patients diagnosed with metastatic castrate-resistant prostate cancer, several new drugs have become available which may provide a clear survival benefit but the optimal choice will have to be made on an individual basis.
Collapse
Affiliation(s)
- Philip Cornford
- Royal Liverpool and Broadgreen Hospitals NHS Trust, Liverpool, UK.
| | - Joaquim Bellmunt
- Bladder Cancer Center, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Michel Bolla
- Department of Radiation Therapy, CHU Grenoble, Grenoble, France
| | | | | | - Tobias Gross
- Department of Urology, University of Bern, Inselspital, Bern, Switzerland
| | - Ann M Henry
- Leeds Cancer Centre, St. James's University Hospital, Leeds, UK
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Thomas B Lam
- Academic Urology Unit, University of Aberdeen, Aberdeen, UK; Department of Urology, Aberdeen Royal Infirmary, Aberdeen, UK
| | | | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Olivier Rouvière
- Hospices Civils de Lyon, Radiology Department, Edouard Herriot Hospital, Lyon, France
| | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Nicolas Mottet
- Department of Urology, University Hospital, St. Etienne, France
| |
Collapse
|
33
|
Mehraein-Ghomi F, Church DR, Schreiber CL, Weichmann AM, Basu HS, Wilding G. Inhibitor of p52 NF-κB subunit and androgen receptor (AR) interaction reduces growth of human prostate cancer cells by abrogating nuclear translocation of p52 and phosphorylated AR(ser81). Genes Cancer 2015; 6:428-44. [PMID: 26622945 PMCID: PMC4633170 DOI: 10.18632/genesandcancer.77] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Accumulating evidence shows that androgen receptor (AR) activation and signaling plays a key role in growth and progression in all stages of prostate cancer, even under low androgen levels or in the absence of androgen in the castration-resistant prostate cancer. Sustained activation of AR under androgen-deprived conditions may be due to its interaction with co-activators, such as p52 NF-κB subunit, and/or an increase in its stability by phosphorylation that delays its degradation. Here we identified a specific inhibitor of AR/p52 interaction, AR/p52-02, via a high throughput screen based on the reconstitution of Gaussia Luciferase. We found that AR/p52-02 markedly inhibited growth of both castration-resistant C4-2 (IC50 ∼6 μM) and parental androgen-dependent LNCaP (IC50 ∼4 μM) human prostate cancer cells under low androgen conditions. Growth inhibition was associated with significantly reduced nuclear p52 levels and DNA binding activity, as well as decreased phosphorylation of AR at serine 81, increased AR ubiquitination, and decreased AR transcriptional activity as indicated by decreased prostate-specific antigen (PSA) mRNA levels in both cell lines. AR/p52-02 also caused a reduction in levels of p21(WAF/CIP1), which is a direct AR targeted gene in that its expression correlates with androgen stimulation and mitogenic proliferation in prostate cancer under physiologic levels of androgen, likely by disrupting the AR signaling axis. The reduced level of cyclinD1 reported previously for this compound may be due to the reduction in nuclear presence and activity of p52, which directly regulates cyclinD1 expression, as well as the reduction in p21(WAF/CIP1), since p21(WAF/CIP1) is reported to stabilize nuclear cyclinD1 in prostate cancer. Overall, the data suggest that specifically inhibiting the interaction of AR with p52 and blocking activity of p52 and pARser81 may be an effective means of reducing castration-resistant prostate cancer cell growth.
Collapse
Affiliation(s)
| | - Dawn R Church
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | | | | | - Hirak S Basu
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - George Wilding
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
34
|
Klotz L, O'Callaghan C, Ding K, Toren P, Dearnaley D, Higano CS, Horwitz E, Malone S, Goldenberg L, Gospodarowicz M, Crook JM. Nadir testosterone within first year of androgen-deprivation therapy (ADT) predicts for time to castration-resistant progression: a secondary analysis of the PR-7 trial of intermittent versus continuous ADT. J Clin Oncol 2015; 33:1151-6. [PMID: 25732157 PMCID: PMC4372851 DOI: 10.1200/jco.2014.58.2973] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Three small retrospective studies have suggested that patients undergoing continuous androgen deprivation (CAD) have superior survival and time to progression if lower castrate levels of testosterone (< 0.7 nmol/L) are achieved. Evidence from prospective large studies has been lacking. PATIENTS AND METHODS The PR-7 study randomly assigned patients experiencing biochemical failure after radiation therapy or surgery plus radiation therapy to CAD or intermittent androgen deprivation. The relationship between testosterone levels in the first year and cause-specific survival (CSS) and time to androgen-independent progression in men in the CAD arm was evaluated using Cox regression. RESULTS There was a significant difference in CSS (P = .015) and time to hormone resistance (P = .02) among those who had first-year minimum nadir testosterone ≤ 0.7, > 0.7 to ≤ 1.7, and ≥ 1.7 nmol/L. Patients with first-year nadir testosterone consistently > 0.7 nmol/L had significantly higher risks of dying as a result of disease (0.7 to 1.7 nmol/L: hazard ratio [HR], 2.08; 95% CI, 1.28 to 3.38; > 1.7 nmol/L: HR, 2.93; 95% CI, 0.70 to 12.30) and developing hormone resistance (0.7 to 1.7 nmol/L: HR, 1.62; 95% CI, 1.20 to 2.18; ≥ 1.7 nmol/L: HR, 1.90; 95% CI, 0.77 to 4.70). Maximum testosterone ≥ 1.7 nmol/L predicted for a higher risk of dying as a result of disease (P = .02). CONCLUSION Low nadir serum testosterone (ie, < 0.7 mmol/L) within the first year of androgen-deprivation therapy correlates with improved CSS and duration of response to androgen deprivation in men being treated for biochemical failure undergoing CAD.
Collapse
Affiliation(s)
- Laurence Klotz
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA.
| | - Chris O'Callaghan
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Keyue Ding
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Paul Toren
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - David Dearnaley
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Celestia S Higano
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Eric Horwitz
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Shawn Malone
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Larry Goldenberg
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Mary Gospodarowicz
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Juanita M Crook
- Laurence Klotz, Sunnybrook Health Sciences Centre, University of Toronto; Mary Gospodarowicz, Princess Margaret Hospital, Toronto; Chris O'Callaghan and Keyue Ding, Queen's University, Kingston; Shawn Malone, Ottawa Regional Cancer Centre, Ottawa, Ontario; Paul Toren, Larry Goldenberg, and Juanita M. Crook, University of British Columbia, Vancouver, British Columbia, Canada; David Dearnaley, Royal Marsden Hospital, London, United Kingdom; Celestia S. Higano, Fox Chase Cancer Center, Philadelphia, PA; and Eric Horwitz, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
35
|
Shiota M, Yokomizo A, Takeuchi A, Imada K, Kiyoshima K, Inokuchi J, Tatsugami K, Naito S. The oncological outcome and validation of Japan Cancer of the Prostate Risk Assessment score among men treated with primary androgen-deprivation therapy. J Cancer Res Clin Oncol 2014; 141:495-503. [DOI: 10.1007/s00432-014-1828-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/08/2014] [Indexed: 12/27/2022]
|
36
|
Nishiyama T. Serum testosterone levels after medical or surgical androgen deprivation: a comprehensive review of the literature. Urol Oncol 2013; 32:38.e17-28. [PMID: 23769268 DOI: 10.1016/j.urolonc.2013.03.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/26/2013] [Accepted: 03/26/2013] [Indexed: 11/15/2022]
Abstract
Androgens and the androgen receptor play a role in the progression of prostate cancer. Androgen deprivation therapy (ADT) is a mainstay in the treatment of metastatic prostate cancer. ADT is expected to reduce serum testosterone levels from a normal level of about 500 to 600 ng/dl (17.3-20.8 nmol) down to castration levels. Traditionally, castration was considered to be achieved if testosterone levels were lowered to a threshold of 50 ng/dl (1.73 nmol/l), a definition determined more by measurement methods derived from the use of old assay methods than by evidence. Serum testosterone levels in three-quarter patients after surgical castration drop to less than 20 ng/dl (0.69 nmol/l). Ineffective suppression of testosterone is currently poorly recognized and may possibly have an effect of prostate cancer mortality. Persistent levels of serum testosterone after castration are mainly derived from adrenal androgens. Furthermore, the arrival of new therapies targeting androgen synthesis and androgen receptor activity has renewed interest on serum testosterone. This review discusses the biosynthetic pathway for androgen synthesis in humans and provides a comprehensive review of serum testosterone levels after surgical or medical castration. This review assesses serum testosterone levels after surgical castration and different pharmacologic castration in patients with prostate cancer under ADT, and ineffective testosterone suppression. The author proposes methods to better lower serum testosterone levels during ADT.
Collapse
Affiliation(s)
- Tsutomu Nishiyama
- Division of Urology, Department of Regenerative and Transplant Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
37
|
Dason S, Allard CB, Tong J, Shayegan B. Defining a new testosterone threshold for medical castration: Results from a prospective cohort series. Can Urol Assoc J 2013; 7:E263-7. [PMID: 23766827 PMCID: PMC3668394 DOI: 10.5489/cuaj.471] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND We seek to determine if testosterone levels below the accepted castration threshold (50 ng/dL) have an impact on time to progression to castrate-resistant prostate cancer (CRPC). METHODS This is a prospective cohort series of patients undergoing androgen deprivation therapy (ADT) with luteinizing hormone-releasing hormone agonist or antagonist at a tertiary centre from 2006 to 2011. Serum testosterone level was assessed every 3 months. Patients with any testosterone >50 ng/dL were excluded. Patients were stratified into groups based on those achieving mean testosterone levels <20 ng/dL and <32 ng/dL. Progression to CRPC was assessed with the Kaplan-Meier method and compared with the log-rank test. RESULTS A total of 32 patients were included in this study. Mean patient follow-up was 25.7 months. Patients with a 9-month serum testosterone <32 ng/dL had a significantly increased time to CRPC compared to patients with testosterone 32 to 50 ng/dL (p = 0.001, median progression-free survival (PFS) 33.1 months [<32 ng/dL] vs. 12.5 months [>32 ng/dL]). Patients with first year mean testosterone <32 ng/dL also had a significantly increased time to CRPC compared to 32 to 50 ng/dL (p = 0.05, median PFS 33.1 months [<32 ng/dL] vs. 12.5 months [32-50 ng/dL]). A testosterone <20 ng/dL compared to 20 to 50 ng/dL did not significantly predict with time to CRPC. CONCLUSION This study supports a lower testosterone threshold to define optimal medical castration (T <32 ng/dL) than the previously accepted standard of 50 ng/dL. Testosterone levels during ADT serve as an early predictor of disease progression and thus should be measured in conjunction with prostate-specific antigen.
Collapse
Affiliation(s)
- Shawn Dason
- Division of Urology, Department of Surgery, McMaster University, Hamilton, ON
| | | | - Justin Tong
- Division of Urology, Department of Surgery, McMaster University, Hamilton, ON
| | - Bobby Shayegan
- Division of Urology, Department of Surgery, McMaster University, Hamilton, ON
| |
Collapse
|
38
|
Rove KO, Debruyne FM, Djavan B, Gomella LG, Koul HK, Lucia MS, Petrylak DP, Shore ND, Stone NN, Crawford ED. Role of Testosterone in Managing Advanced Prostate Cancer. Urology 2012; 80:754-62. [DOI: 10.1016/j.urology.2012.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 04/19/2012] [Accepted: 05/05/2012] [Indexed: 10/28/2022]
|
39
|
Schröder F, Crawford ED, Axcrona K, Payne H, Keane TE. Androgen deprivation therapy: past, present and future. BJU Int 2012; 109 Suppl 6:1-12. [PMID: 22672120 DOI: 10.1111/j.1464-410x.2012.11215.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Since Huggins and Hodges demonstrated the responsiveness of prostate cancer to androgen deprivation therapy (ADT), androgen-suppressing strategies have formed the cornerstone of management of advanced prostate cancer. Approaches to ADT have included orchidectomy, oestrogens, luteinizing hormone-releasing hormone (LHRH) agonists, anti-androgens and more recently the gonadotrophin-releasing hormone antagonists. The most extensively studied antagonist, degarelix, avoids the testosterone surge and clinical flare associated with LHRH agonists, offering more rapid PSA and testosterone suppression, improved testosterone control and improved PSA progression-free survival compared with agonists. The clinical profile of degarelix appears to make it a particularly suitable therapeutic option for certain subgroups of patients, including those with metastatic disease, high baseline PSA (>20 ng/mL) and highly symptomatic disease. As well as forming the mainstay of treatment for advanced prostate cancer, ADT is increasingly used in earlier disease stages. While data from clinical trials support the use of ADT neoadjuvant/adjuvant to radiotherapy for locally advanced or high-risk localized prostate cancer, it remains to be established whether specific ADT classes/agents provide particular benefits in this clinical setting.
Collapse
Affiliation(s)
- F Schröder
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
40
|
Schweizer MT, Antonarakis ES. Abiraterone and other novel androgen-directed strategies for the treatment of prostate cancer: a new era of hormonal therapies is born. Ther Adv Urol 2012; 4:167-78. [PMID: 22852027 PMCID: PMC3398601 DOI: 10.1177/1756287212452196] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The number of life-prolonging therapies proven effective in the treatment of metastatic castrate-resistant prostate cancer (CRPC) has been limited until recently. In the past 2 years several such therapies have come to market. In 2010, the autologous immunotherapy sipuleucel-T and the next-generation taxane cabazitaxel were approved in this setting. However, abundant evidence has shown that CRPC growth continues to be driven through androgen-dependent signaling. Both of these drugs fail to take advantage of this targetable oncogenic pathway. Potent specific inhibitors of cytochrome P450-17 have been engineered with the aim of suppressing androgen synthesis beyond that seen with the luteinizing hormone-releasing hormone agonists/antagonists. Abiraterone acetate was developed by rational design based on a pregnenolone parent structure. Its approval by the US Food and Drug Administration (FDA) was granted in 2011 based on phase III data demonstrating an overall survival advantage compared with placebo. More recently, other drugs that act along the androgen signaling pathway, such as orteronel (TAK-700), galeterone (TOK-001), enzalutamide (MDV3100) and ARN-509, have shown promise in clinical trials. Some of these are expected to gain FDA approval in the near future. Here, we review abiraterone and other novel androgen-directed therapeutic strategies for the management of advanced prostate cancer.
Collapse
Affiliation(s)
- Michael T Schweizer
- Prostate Cancer Research Program, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | |
Collapse
|
41
|
Reis LO. Variations of serum testosterone levels in prostate cancer patients under LH-releasing hormone therapy: an open question. Endocr Relat Cancer 2012; 19:R93-8. [PMID: 22399012 DOI: 10.1530/erc-12-0040] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The hypothesis 'the lower the better when achieving castration levels of testosterone' is based on the data from second-line hormonal manipulation and its molecular basis, and on better oncological results reported for lower castration levels in prostate cancer (PCa) patients, including those achieved with maximal androgen blockade. In this regard, the equivalence of surgical and different pharmacological castrations has been controversial. The modified amino acid structure that makes LH-releasing hormone (LHRH) analogs more potent than LHRH, and the method of delivering the analogs impacts on bioavailibility and potentially causes differences in androgen levels and in its final oncological efficacy. In addition to this, there is a myriad of circumstances, such as those related to ethnic variations and co-morbidities, which uniquely impact on the pharmacological approach in a highly heterogeneous population of castration-resistant prostate cancer (CRPC) patients. Ineffective testosterone suppression through hormonal escape is currently poorly recognized and may result in increased PCa mortality. Until now, the optimal serum testosterone level in patients under castration, and the impact of its variations in patients under LHRH therapy, remain open questions and have been merged to a broad spectra of patients who are highly heterogeneous. This heterogeneity relates to a number of mechanisms regarding response to treatment, which influences the biology of the relapsing tumor and the sensitivity to subsequent therapies in the individual patient. The rationale to achieve testosterone levels below 20-50 ng/dl warrant further investigation as these levels have recently rescued CRPC patients. In the last few years and months, important advancements in prostate cancer treatment have been achieved. Nevertheless, these advances are measured in a few months of additional survival and under high costs, not available to most of the world population, compared with the benefits of hormonal manipulation that are measured in years, there is a huge potential for accessible and durable effect expansion and optimization of treatment, particularly with the current tendency of a more individual approach.
Collapse
Affiliation(s)
- Leonardo Oliveira Reis
- Department of Surgery (Urology), Faculty of Medical Sciences, University of Campinas (Unicamp), Rua Tessália Vieira de Camargo, 126 Cidade Universitária Zeferino Vaz, Campinas, São Paulo CEP 13083-887, Brazil.
| |
Collapse
|
42
|
Pickles T, Hamm J, Morris WJ, Schreiber WE, Tyldesley S. Incomplete testosterone suppression with luteinizing hormone-releasing hormone agonists: does it happen and does it matter? BJU Int 2012; 110:E500-7. [PMID: 22564197 DOI: 10.1111/j.1464-410x.2012.11190.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
UNLABELLED What's known on the subject? and What does the study add? Previous reports, with small numbers of patients, have described the problem of incomplete testosterone suppression (>1.1 or 1.7 nmol/L) with LHRH agonists. Various predisposing factors have been suggested: different drug agents and patient factors such as age, pretreatment testosterone levels and weight. Such incomplete testosterone suppression has been shown in one small report to be associated with increased PSA failure rates and in another report in those with metastases, with worse survival. This study used testosterone assays that are more accurate at low levels than those used in most previous reports in a large dataset of 2196 men, and confirmed incomplete testosterone suppression (breakthrough) rates >1.7 nmol/L of 3.4% and >1.1 nmol/L of 6.6%. We showed that younger age was strongly associated with the risk of breakthrough, with a minor effect of increasing body mass index. Repeated breakthroughs were more common (16%) in those who had already had one breakthrough. Interim measures of cancer control (PSA kinetics during LHRH therapy) were inferior in those with a breakthrough, and those with breakthroughs between 1.1 and 1.7 nmol/L had worse long-term biochemical control rates. OBJECTIVES • To describe breakthrough rates above castrate levels of testosterone, in a population-based series of men undergoing adjuvant luteinizing hormone-releasing hormone (LHRH) agonist therapy with curative radiation therapy. • To explore the predisposing factors for such breakthroughs and their impact on subsequent outcomes. PATIENTS AND METHODS • All men treated for prostate cancer between 1998 and 2007 with curative radiation in the province of British Columbia, Canada were potentially eligible (n= 11752). Of these, 2196 fulfilled the eligibility criteria. • Serial testosterone measurements were obtained during continuous LHRH therapy. • Breakthrough rates >1.1 nmol/L and >1.7 nmol/L were calculated for each LHRH injection and for each patient course. • Predisposing factors were identified, and early surrogates of oncological outcome (neoadjuvant nadir and post-treatment nadir) were determined. RESULTS • The risk of a breakthrough >1.1 nmol/L was 6.6%, and >1.7 nmol/L was 3.4% per patient course and 5.4% and 2.2% per LHRH injection (inclusive ranges). • Repeated breakthroughs occurred in 16% of patients. • Younger men were more liable to breakthroughs (P < 0.001). • Early PSA kinetic surrogates of cancer control were inferior in those with breakthroughs. • Neither overall biochemical non-evidence of disease (bNED) nor survival were compromised, although subgroup analysis showed inferior 5-year bNED in those with breakthroughs of 1.1-1.7 nmol/L vs those without (58% vs 73%, respectively; P= 0.048). CONCLUSIONS • Breakthroughs with LHRH agonists occur occasionally per injection, but occur commonly per patient course of treatment, and adversely affect early surrogate measures of outcome. • The monitoring of testosterone levels during therapy is therefore advised.
Collapse
Affiliation(s)
- Tom Pickles
- Radiation Programme, BC Cancer Agency, and Department of Radiotherapy and Developmental Radiotherapeutics, University of British Columbia, Vancouver, Canada.
| | | | | | | | | |
Collapse
|
43
|
Djavan B, Eastham J, Gomella L, Tombal B, Taneja S, Dianat SS, Kazzazi A, Shore N, Abrahamsson PA, Cheetham P, Moul J, Lepor H, Crawford ED. Testosterone in prostate cancer: the Bethesda consensus. BJU Int 2011; 110:344-52. [PMID: 22129242 DOI: 10.1111/j.1464-410x.2011.10719.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
OBJECTIVE • Androgen stimulation of prostate cancer (PCa) cells has been extensively studied. The increasing trend of using serum testosterone as an absolute surrogate for castration state means that the diagnostic measurement of testosterone and the values potentially influencing prognosis must be better understood. This is especially important when PCa progresses from an endocrine to an intracrine status. PATIENTS AND METHODS • We performed a literature review using the MEDLINE database for publications on: (i) hormonal changes with androgen deprivation therapy (ADT); (ii) monitoring hormonal therapy with testosterone measurement; (iii) the efficacy of intermittent androgen deprivation (IAD) compared with continuous androgen deprivation; (iv) the underlying mechanisms of castration-resistance; and (v) novel treatments for castration-resistant PCa (CRPCa). RESULTS • The optimum serum castration levels to be achieved with ADT are still debated. Recently, the 50 ng/dL threshold has been questioned because of reports indicating worse outcomes when levels between 20 and 50 ng/dL were studied. Instead, a 20 ng/dL threshold for serum testosterone after ADT in patients with advanced prostate cancer was recommended. CONCLUSION • Understanding the mechanisms of androgen biosynthesis relating to PCa as well as prognostic implications might achieve a consensus regarding the role of ADT for both the androgen-sensitive and -insensitive disease state.
Collapse
Affiliation(s)
- Bob Djavan
- Department of Urology, New York University School of Medicine, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Development and clinical utility of abiraterone acetate as an androgen synthesis inhibitor. Clin Pharmacol Ther 2011; 91:101-8. [PMID: 22130117 DOI: 10.1038/clpt.2011.275] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In April 2011, abiraterone acetate (AA) was approved by the US Food and Drug Administration (FDA) for the treatment of metastatic castration-resistant prostate cancer (CRPC) after chemotherapy. The development of AA is the direct result of our increased understanding of the intricacies of the androgen receptor (AR) pathway and its natural evolution in prostate cancer cells over the course of treatment. In this paper we review the biology of the AR and how it led to the rational design of AA. We also examine the clinical development of AA, its current use, and questions to be addressed for future development.
Collapse
|
45
|
Ryan CJ, Tindall DJ. Androgen receptor rediscovered: the new biology and targeting the androgen receptor therapeutically. J Clin Oncol 2011; 29:3651-8. [PMID: 21859989 DOI: 10.1200/jco.2011.35.2005] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Discoveries over the past decade suggest that castration-resistant prostate cancer (CRPC) is sensitive, but not resistant to, further manipulation of the androgen-androgen receptor (AR) axis. Several new therapies that target this axis have demonstrated clinical activity. In this article, preclinical and clinical findings occurring in the field of AR-targeted therapies are reviewed. Reviews of scientific and clinical development are divided into those occurring prereceptor (androgen production and conversion) and at the level of the receptor (AR aberrations and therapies targeting AR directly). Intracrine androgen production and AR amplification, among others, are among the principal aberrancies driving CRPC growth. Phase III data with abiraterone acetate and phase II data with MDV-3100, along with other similar therapies, confirm for the clinician that the scientific findings related to persistent AR signaling in a castrate milieu can be harnessed to produce significant clinical benefit for patients with the disease. Studies aimed at optimizing the timing of their use and exploring the mechanisms of resistance to these therapies are under way. The clinical success of therapies that directly target androgen synthesis as well as the most common aberrancies of the AR confirm that prostate cancer retains dependence on AR signaling, even in the castrate state.
Collapse
Affiliation(s)
- Charles J Ryan
- UCSF Helen Diller Family Comprehensive Cancer Center, 1600 Divisadero St, San Francisco, CA 94115, USA.
| | | |
Collapse
|
46
|
Schulman CC, Irani J, Morote J, Schalken JA, Montorsi F, Chlosta PL, Heidenreich A. Androgen-Deprivation Therapy in Prostate Cancer: A European Expert Panel Review. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.eursup.2010.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Takizawa I, Nishiyama T, Hara N, Isahaya E, Hoshii T, Takahashi K. Serum prostate-specific antigen levels reflect the androgen milieu in patients with localized prostate cancer receiving androgen deprivation therapy: Tumor malignant potential and androgen milieu. Prostate 2010; 70:1395-401. [PMID: 20687212 DOI: 10.1002/pros.21174] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Although androgen deprivation therapy (ADT) has a marked impact on the androgen milieu in vivo and outcomes of prostate cancer (PCa), it remains unclear which parameters reflect the androgen milieu during ADT or whether the milieu is associated with PCa aggressiveness. METHODS Seventy-two patients with localized PCa were prospectively studied based on their blood samples before and after ADT for 6 months. Serum androgens and related values were measured. RESULTS Before ADT, there was no correlation between the serum prostate-specific antigen (PSA) and androgen levels. After ADT, the serum PSA levels were correlated with each level of serum testosterone, dihydrotestosterone, androstenedione, dehydroepiandrosterone-sulfate (DHEA-S), and 3alpha-diol G (P < 0.010 in all). Before ADT, patients with Gleason score of > or = 8 were likely to have lower serum testosterone levels than those with Gleason score of < or = 6 (P = 0.058). After ADT, conversely, the testosterone levels in patients with Gleason score of > or = 8 appeared to be higher than in those with Gleason score of < or = 6 (P = 0.060). The serum DHEA-S level was correlated with Gleason score before and after ADT (P = 0.050 and P = 0.040, respectively). CONCLUSIONS The serum PSA levels well reflect the androgen milieu in localized PCa patients receiving ADT, which can be explained by the Saturation Model and disease control. The androgen milieu in men with high Gleason score PCa is probably less affected by conventional ADT than that in men with low score cancer, which was suggested to be associated with adrenal androgen levels.
Collapse
Affiliation(s)
- Itsuhiro Takizawa
- Division of Urology, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Schulman CC, Irani J, Morote J, Schalken JA, Montorsi F, Chlosta PL, Heidenreich A. Testosterone Measurement in Patients with Prostate Cancer. Eur Urol 2010; 58:65-74. [DOI: 10.1016/j.eururo.2010.04.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 04/06/2010] [Indexed: 10/19/2022]
|
49
|
Current Opinion in Endocrinology, Diabetes & Obesity. Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:293-312. [PMID: 20418721 DOI: 10.1097/med.0b013e328339f31e] [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] [Indexed: 11/25/2022]
|
50
|
INDIVIDUAL VARIATIONS OF SERUM TESTOSTERONE IN PATIENTS WITH PROSTATE CANCER RECEIVING ANDROGEN-DEPRIVATION THERAPY. BJU Int 2009; 104:124; author reply 124-5. [DOI: 10.1111/j.1464-410x.2009.08722_1.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|