1
|
Wang T, Wang X, Ding G, Liu H, Ma X, Ma J, Cui Y, Wu J. Efficacy and safety evaluation of androgen deprivation therapy-based combinations for metastatic castration-sensitive prostate cancer: a systematic review and network meta-analysis. Br J Cancer 2024; 131:1363-1377. [PMID: 39223303 PMCID: PMC11479264 DOI: 10.1038/s41416-024-02823-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND This systematic review and network meta-analysis aimed to assess the comparative effectiveness and safety profiles of current combination therapies based on androgen deprivation therapy (ADT) for the heterogeneous population of individuals with metastatic castration-sensitive prostate cancer (mCSPC). METHODS We retrieved pertinent literature from PubMed, EMBASE, the Cochrane Library, ClinicalTrials.gov, and international conference databases. The study was registered in the Prospective Register of Systematic Reviews (CRD42023453853) for transparency. RESULTS Our analysis included 20 RCTs involving 14,995 patients, evaluating 15 ADT-based combinations, including systemic therapies, radiotherapy and surgery. In the overall population, the darolutamide triplet (DARO + docetaxel + ADT) demonstrated comparable overall survival (OS) benefits to prostatectomy/radical local therapy (RLT) plus ADT (hazard ratio [HR], 0.82; 95% confidence interval [CI], 0.43-1.57). Additionally, the enzalutamide (ENZ) triplet (ENZ + DOC + ADT) appeared to confer the best progression-free survival (HR, 0.34; 95% CI: 0.27-0.43). Subgroup analysis based on metastatic burden indicated that RLT plus ADT had the best OS performance in patients with low burden, while the DARO triplet was associated with the best OS in patients with high burden. Regarding adverse events (AEs), the addition of certain androgen receptor pathway inhibitor (ARPI) agents to ADT led to an increased incidence of severe AEs, while the addition of DOC to the ARPI doublet did not appear to elevate the exposure-adjusted incidence rates. CONCLUSIONS Our findings suggest that combined treatments result in better survival outcomes than does ADT alone. In the current landscape of systemic therapy, the significance of local therapy should not be underestimated, and therapeutic decisions should be tailored with meticulous consideration of clinical heterogeneity among patients.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Xiaoyu Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guixin Ding
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Hongquan Liu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Xiaohong Ma
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Jian Ma
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Yuanshan Cui
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China.
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China.
| |
Collapse
|
2
|
Deek MP, Sutera P, Jing Y, Gao R, Rothman E, Day H, Chang D, Dirix P, Armstrong AJ, Campbell B, Lopez Campos F, Berenguer M, Ramotar M, Conde-Moreno A, Berlin A, Bosetti DG, Corcoran N, Koontz B, Mercier C, Siva S, Pryor D, Ost P, Huynh MA, Kroeze S, Stish B, Kiess A, Trock B, Tran PT, Gillessen S, Sweeney C. Multi-institutional Analysis of Metastasis-directed Therapy with or Without Androgen Deprivation Therapy in Oligometastatic Castration-sensitive Prostate Cancer. Eur Urol Oncol 2024:S2588-9311(24)00086-5. [PMID: 38570239 DOI: 10.1016/j.euo.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/10/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Metastasis-directed therapy (MDT) is increasingly being used in oligometastatic castration-sensitive prostate cancer (omCSPC). However, it is currently unclear how to optimally integrate MDT with the standard of care of systemic hormonal therapy. OBJECTIVE To report long-term outcomes of MDT alone versus MDT and a defined course of androgen deprivation therapy (ADT) in omCSPC. DESIGN, SETTING, AND PARTICIPANTS Here, a multicenter, international retrospective cohort of omCSPC as defined by conventional imaging was reported. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Biochemical progression-free survival (bPFS), distant progression-free survival (dPFS), and combined biochemical or distant progression-free survival (cPFS) were evaluated with Kaplan-Meier and multivariable Cox proportional hazard regression models. RESULTS AND LIMITATIONS A total of 263 patients were included, 105 with MDT + ADT and 158 with MDT alone. The majority of patients had metachronous disease (90.5%). Five-year bPFS, dPFS, and cPFS were, respectively, 24%, 41%, and 19% in patients treated with MDT + ADT and 11% (hazard ratio [HR] 0.48, 95% confidence interval [CI] 0.36-0.64), 29% (HR 0.56, 95% CI 0.40-0.78), and 9% (HR 0.50, 95% CI 0.38-0.67) in patients treated with MDT alone. On a multivariable analysis adjusting for pretreatment variables, the use of ADT was associated with improved bPFS (HR 0.43, p < 0.001), dPFS (HR 0.45, p = 0.002), and cPFS (HR 0.44, p < 0.001). CONCLUSIONS In this large multi-institutional report, the addition of concurrent ADT to MDT appears to improve time to prostate-specific antigen progression and distant recurrence, noting that about 10% patients had durable control with MDT alone. Ongoing phase 3 studies will help further define treatment options for omCSPC. PATIENT SUMMARY Here, we report a large retrospective review evaluating the outcomes of metastasis-directed therapy with or without a limited course of androgen deprivation for patients with oligometastatic castration-sensitive prostate cancer. This international multi-institutional review demonstrates that the addition of androgen deprivation therapy to metastasis-directed therapy (MDT) improves progression-free survival. While a proportion of patients appear to have long-term disease control with MDT alone, further work in biomarker discovery is required to better identify which patients would be appropriate for de-escalated therapy.
Collapse
Affiliation(s)
- Matthew P Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuezhou Jing
- The James Buchanan Brady Urological Institute of Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Gao
- Department of Radiation Oncology, The Mayo Clinic, Rochester, MN, USA
| | - Emily Rothman
- Department of Radiation Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Heather Day
- Department of Radiation Oncology, Australian Prostate Cancer Research Center, Queensland, Australia
| | - David Chang
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - Piet Dirix
- Department of Radiation-Oncology, GasthuisZusters Antwerp (GZA) 'Sisters of the Hospital', Antwerp, Belgium
| | - Andrew J Armstrong
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute Center for Prostate and Urologic Cancer, Duke University Medical Center, Durham, NC, USA
| | - Bethany Campbell
- Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | - Miguel Berenguer
- Radiation Oncology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Matthew Ramotar
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Antonio Conde-Moreno
- Radiation Oncology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Alejandro Berlin
- Radiation Oncology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Davide Giovanni Bosetti
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Niall Corcoran
- Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | - Carole Mercier
- Department of Radiation-Oncology, GasthuisZusters Antwerp (GZA) 'Sisters of the Hospital', Antwerp, Belgium
| | - Shankar Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - David Pryor
- Department of Radiation Oncology, Australian Prostate Cancer Research Center, Queensland, Australia
| | - Piet Ost
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Mai Anh Huynh
- Department of Radiation Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Stephanie Kroeze
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Bradley Stish
- Department of Radiation Oncology, The Mayo Clinic, Rochester, MN, USA
| | - Ana Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruce Trock
- The James Buchanan Brady Urological Institute of Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Christopher Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
3
|
Sutera P, Shetty AC, Song Y, Hodges T, Hoang T, Rana Z, Pienta K, Feng F, Song DY, DeWeese T, Gillessen S, Sweeney C, James N, Attard G, Deek M, Tran PT. Identification of a Predictive Genomic Biomarker for Prostate-directed Therapy in Synchronous Low-volume Metastatic Castration-sensitive Prostate Cancer. Eur Urol Oncol 2024; 7:241-247. [PMID: 37558543 PMCID: PMC10850431 DOI: 10.1016/j.euo.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/11/2023] [Accepted: 07/26/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Standard of care management for synchronous metastatic castration-sensitive prostate cancer (mCSPC) includes androgen deprivation therapy with a second-generation antiandrogen therapy and/or docetaxel. Recently, randomized data have demonstrated that prostate-directed therapy (PDT) is associated with an improvement in overall survival (OS) among patients with low-volume metastatic disease. Tumor genomics represents an additional dimension to define the clinical trajectory of patients with mCSPC. OBJECTIVE To evaluate a high-risk (HiRi) genomic signature to predict the benefit from PDT. DESIGN, SETTING, AND PARTICIPANTS We performed a single-institution retrospective review of men with synchronous low-volume mCSPC who underwent DNA panel sequencing of their tumor. Patients were classified according to the presence of HiRi mutation including pathogenic mutations in TP53, ATM, BRCA1, BRCA2, or Rb1. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint was to determine the effect of PDT on OS in patients with and without a HiRi mutation. A survival analysis was performed with the Kaplan-Meier method compared with log-rank test and multivariable Cox regression. The interaction between HiRi mutation and PDT was evaluated. RESULTS AND LIMITATIONS A total of 101 patients with synchronous low-volume CSPC were included with a median follow-up of 44 mo. Approximately half of patients were found to have a HiRi pathogenic mutation (49%). Patients with HiRi mutations demonstrated median OS of 73 versus 66.8 mo (p = 0.3) for no PDT versus PDT. Conversely, patients without a HiRi mutation demonstrated a significant improvement in OS of 60 versus 105.3 mo (p < 0.001) for no PDT versus PDT. The p value for interaction for OS between PDT and HiRi mutation was statistically significant (p < 0.001). Limitations include the retrospective nature of the study. CONCLUSIONS Here, we have identified a HiRi genomic biomarker that appears predictive for the lack of benefit from PDT in men with synchronous low-volume mCSPC. Further work validating these results is warranted. PATIENT SUMMARY In this report, we evaluated a high-risk genomic biomarker to predict the benefit from prostate-directed therapy for men with synchronous low-volume metastatic castration-sensitive prostate cancer. We found that men without a high-risk mutation appear to experience a greater clinical benefit from prostate-directed therapy than those with a high-risk mutation.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Theresa Hodges
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tung Hoang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biochemistry and Molecular Biology, Johns Hopkins University, School of Public Health, Baltimore, MD, USA
| | - Zaker Rana
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kenneth Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Felix Feng
- Departments of Medicine, Urology and Radiation Oncology, UCSF, San Francisco, CA, USA
| | - Daniel Y Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Theodore DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Silke Gillessen
- Istituto Oncologico della Svizzera Italiana, Bellinzona, Switzerland
| | - Christopher Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia
| | - Nicholas James
- The Royal Marsden Hospital NHS Foundation Trust and The Institute of Cancer Research, London, UK
| | - Gerhardt Attard
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Matthew Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Kishan AU, Siva S, Hofman MS, Nagarajah J, Kiess AP, Tran P, Calais J. The Potential Contribution of Radiopharmaceutical Therapies in Managing Oligometastatic Disease. J Nucl Med 2024:jnumed.123.266772. [PMID: 38360048 DOI: 10.2967/jnumed.123.266772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/23/2024] [Indexed: 02/17/2024] Open
Abstract
There is a growing understanding of the oligometastatic disease state, characterized by the presence of 5 or fewer lesions. Advanced molecular imaging techniques, such as prostate-specific membrane antigen PET, refines the ability to detect oligometastatic recurrences (oligorecurrences) early. These developments have led to the exploration of metastasis-directed therapy (MDT) in oligorecurrent disease as an alternative to or as a means of delaying systemic therapy. Unfortunately, MDT often does not provide a durable cure, and progression-particularly progression in multiple new areas-remains a concern. Simultaneously, developments in radioligand therapy (RLT) have led to studies showing overall survival benefits with α-emitting and β-emitting RLT in advanced, high-volume, metastatic castration-resistant prostate cancer. The success of RLT in late-stage disease suggests that earlier use in the disease spectrum may be impactful. Specifically, integration of RLT with MDT might reduce progression, including polymetastatic progression, in the setting of oligorecurrent disease.
Collapse
Affiliation(s)
- Amar U Kishan
- Department of Radiation Oncology, UCLA, Los Angeles, California;
- Department of Urology, UCLA, Los Angeles, California
| | - Shankar Siva
- Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Ana P Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Phuoc Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| |
Collapse
|
5
|
Hoang T, Sutera P, Nguyen T, Chang J, Jagtap S, Song Y, Shetty AC, Chowdhury DD, Chan A, Carrieri FA, Hathout L, Ennis R, Jabbour SK, Parikh R, Molitoris J, Song DY, DeWeese T, Marchionni L, Ren L, Sawant A, Simone N, Lafargue A, Van Der Eecken K, Bunz F, Ost P, Tran PT, Deek MP. TP53 structure-function relationships in metastatic castrate-sensitive prostate cancer and the impact of APR-246 treatment. Prostate 2024; 84:87-99. [PMID: 37812042 DOI: 10.1002/pros.24629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE Despite well-informed work in several malignancies, the phenotypic effects of TP53 mutations in metastatic castration-sensitive prostate cancer (mCSPC) progression and metastasis are not clear. We characterized the structure-function and clinical impact of TP53 mutations in mCSPC. PATIENTS AND METHODS We performed an international retrospective review of men with mCSPC who underwent next-generation sequencing and were stratified according to TP53 mutational status and metastatic burden. Clinical outcomes included radiographic progression-free survival (rPFS) and overall survival (OS) evaluated with Kaplan-Meier and multivariable Cox regression. We also utilized isogenic cancer cell lines to assess the effect of TP53 mutations and APR-246 treatment on migration, invasion, colony formation in vitro, and tumor growth in vivo. Preclinical experimental observations were compared using t-tests and ANOVA. RESULTS Dominant-negative (DN) TP53 mutations were enriched in patients with synchronous (vs. metachronous) (20.7% vs. 6.3%, p < 0.01) and polymetastatic (vs. oligometastatic) (14.4% vs. 7.9%, p < 0.01) disease. On multivariable analysis, DN mutations were associated with worse rPFS (hazards ratio [HR] = 1.97, 95% confidence interval [CI]: 1.31-2.98) and overall survival [OS] (HR = 2.05, 95% CI: 1.14-3.68) compared to TP53 wild type (WT). In vitro, 22Rv1 TP53 R175H cells possessed stronger migration, invasion, colony formation ability, and cellular movement pathway enrichment in RNA sequencing analysis compared to 22Rv1 TP53 WT cells. Treatment with APR-246 reversed the effects of TP53 mutations in vitro and inhibited 22Rv1 TP53 R175H tumor growth in vivo in a dosage-dependent manner. CONCLUSIONS DN TP53 mutations correlated with worse prognosis in prostate cancer patients and higher metastatic potential, which could be counteracted by APR-246 treatment suggesting a potential future therapeutic avenue.
Collapse
Affiliation(s)
- Tung Hoang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins University School of Public Health, Baltimore, Maryland, USA
| | - Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Triet Nguyen
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins University School of Public Health, Baltimore, Maryland, USA
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Jinhee Chang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Shreya Jagtap
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Yang Song
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Amol C Shetty
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dipanwita D Chowdhury
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Aaron Chan
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Francesca A Carrieri
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lara Hathout
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Ronald Ennis
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Rahul Parikh
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Jason Molitoris
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Daniel Y Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Theodore DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Lei Ren
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Amit Sawant
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Nicole Simone
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Audrey Lafargue
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
| | - Kim Van Der Eecken
- Department of Pathology, Ghent University Hospital, Cancer Research Institute (CRIG), Ghent, Belgium
| | - Fred Bunz
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, Antwerp, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Phuoc T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiation Oncology, Division of Translational Radiation Sciences, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, James Buchanan Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew P Deek
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
6
|
Bratic Hench I, Roma L, Conticelli F, Bubendorf L, Calgua B, Le Magnen C, Piscuoglio S, Rubin MA, Chirindel A, Nicolas GP, Vlajnic T, Zellweger T, Templeton AJ, Stenner F, Ruiz C, Rentsch C, Bubendorf L. Cell-Free DNA Genomic Profiling and Its Clinical Implementation in Advanced Prostate Cancer. Cancers (Basel) 2023; 16:45. [PMID: 38201475 PMCID: PMC10778564 DOI: 10.3390/cancers16010045] [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: 11/10/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Most men with prostate cancer (PCa), despite potentially curable localized disease at initial diagnosis, progress to metastatic disease. Despite numerous treatment options, choosing the optimal treatment for individual patients remains challenging. Biomarkers guiding treatment sequences in an advanced setting are lacking. To estimate the diagnostic potential of liquid biopsies in guiding personalized treatment of PCa, we evaluated the utility of a custom-targeted next-generation sequencing (NGS) panel based on the AmpliSeq HD Technology. Ultra-deep sequencing on plasma circulating free DNA (cfDNA) samples of 40 metastatic castration-resistant PCa (mCRPC) and 28 metastatic hormone-naive PCa (mCSPC) was performed. CfDNA somatic mutations were detected in 48/68 (71%) patients. Of those 68 patients, 42 had matched tumor and cfDNA samples. In 21/42 (50%) patients, mutations from the primary tumor tissue were detected in the plasma cfDNA. In 7/42 (17%) patients, mutations found in the primary tumor were not detected in the cfDNA. Mutations from primary tumors were detected in all tested mCRPC patients (17/17), but only in 4/11 with mCSPC. AR amplifications were detected in 12/39 (31%) mCRPC patients. These results indicate that our targeted NGS approach has high sensitivity and specificity for detecting clinically relevant mutations in PCa.
Collapse
Affiliation(s)
- Ivana Bratic Hench
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Luca Roma
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Floriana Conticelli
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Lenard Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Byron Calgua
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Clémentine Le Magnen
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Department of Urology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Salvatore Piscuoglio
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Mark A. Rubin
- Precision Oncology Laboratory, Department for Biomedical Research, Bern Center for Precision Medicine, 3008 Bern, Switzerland
- Bern Center for Precision Medicine, Inselspital, Bern University Hospital, University of Bern, 3008 Bern, Switzerland
| | - Alin Chirindel
- Division of Nuclear Medicine, Department of Theragnostics, University Hospital Basel, 4031 Basel, Switzerland
| | - Guillaume P. Nicolas
- Division of Nuclear Medicine, Department of Theragnostics, University Hospital Basel, 4031 Basel, Switzerland
| | - Tatjana Vlajnic
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | | | - Arnoud J. Templeton
- St. Claraspital, 4058 Basel, Switzerland
- St. Clara Research, Basel and Faculty of Medicine, University Basel, 4058 Basel, Switzerland
| | - Frank Stenner
- Division of Oncology, University Hospital Basel, 4031 Basel, Switzerland
| | - Christian Ruiz
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Cyrill Rentsch
- Department of Urology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
7
|
Sutera PA, Shetty AC, Hakansson A, Van der Eecken K, Song Y, Liu Y, Chang J, Fonteyne V, Mendes AA, Lumen N, Delrue L, Verbeke S, De Man K, Rana Z, Hodges T, Hamid A, Roberts N, Song DY, Pienta K, Ross AE, Feng F, Joniau S, Spratt D, Gillessen S, Attard G, James ND, Lotan T, Davicioni E, Sweeney C, Tran PT, Deek MP, Ost P. Transcriptomic and clinical heterogeneity of metastatic disease timing within metastatic castration-sensitive prostate cancer. Ann Oncol 2023; 34:605-614. [PMID: 37164128 PMCID: PMC10330666 DOI: 10.1016/j.annonc.2023.04.515] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Metastatic castration-sensitive prostate cancer (mCSPC) is commonly classified into high- and low-volume subgroups which have demonstrated differential biology, prognosis, and response to therapy. Timing of metastasis has similarly demonstrated differences in clinical outcomes; however, less is known about any underlying biologic differences between these disease states. Herein, we aim to compare transcriptomic differences between synchronous and metachronous mCSPC and identify any differential responses to therapy. PATIENTS AND METHODS We performed an international multi-institutional retrospective review of men with mCSPC who completed RNA expression profiling evaluation of their primary tumor. Patients were stratified according to disease timing (synchronous versus metachronous). The primary endpoint was to identify differences in transcriptomic profiles between disease timing. The median transcriptomic scores between groups were compared with the Mann-Whitney U test. Secondary analyses included determining clinical and transcriptomic variables associated with overall survival (OS) from the time of metastasis. Survival analysis was carried out with the Kaplan-Meier method and multivariable Cox regression. RESULTS A total of 252 patients were included with a median follow-up of 39.6 months. Patients with synchronous disease experienced worse 5-year OS (39% versus 79%; P < 0.01) and demonstrated lower median androgen receptor (AR) activity (11.78 versus 12.64; P < 0.01) and hallmark androgen response (HAR; 3.15 versus 3.32; P < 0.01). Multivariable Cox regression identified only high-volume disease [hazard ratio (HR) = 4.97, 95% confidence interval (CI) 2.71-9.10; P < 0.01] and HAR score (HR = 0.51, 95% CI 0.28-0.88; P = 0.02) significantly associated with OS. Finally, patients with synchronous (HR = 0.47, 95% CI 0.30-0.72; P < 0.01) but not metachronous (HR = 1.37, 95% CI 0.50-3.92; P = 0.56) disease were found to have better OS with AR and non-AR combination therapy as compared with monotherapy (P value for interaction = 0.05). CONCLUSIONS We have demonstrated a potential biologic difference between metastatic timing of mCSPC. Specifically, for patients with low-volume disease, those with metachronous low-volume disease have a more hormone-dependent transcriptional profile and exhibit a better prognosis than synchronous low-volume disease.
Collapse
Affiliation(s)
- P A Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - A C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | | | - K Van der Eecken
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Y Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | | | - J Chang
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | - V Fonteyne
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - A A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - N Lumen
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - L Delrue
- Department of Radiology, Ghent University Hospital, Ghent, Belgium
| | - S Verbeke
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - K De Man
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Z Rana
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | - T Hodges
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | - A Hamid
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - N Roberts
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, USA
| | - D Y Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA; James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - K Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA; James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - A E Ross
- Department of Urology, Northwestern University, Chicago, USA
| | - F Feng
- Department of Medicine, UCSF, San Francisco, USA; Department of Urology, UCSF, San Francisco, USA; Department of Radiation Oncology, UCSF, San Francisco, USA
| | - S Joniau
- Department of Urology, Catholic University Leuven, Leuven, Belgium
| | - D Spratt
- Department of Radiation Oncology, University Hospitals, Cleveland, USA
| | - S Gillessen
- Istituto Oncologico della Svizzera Italiana, Bellinzona, Switzerland
| | - G Attard
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - N D James
- The Royal Marsden Hospital NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - T Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - C Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia
| | - P T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, USA; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, USA
| | - M P Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, USA.
| | - P Ost
- Department of Radiation Oncology, Iridium Network, Antwerp, Belgium; Department of Human Structure and Repair, Ghent University, Ghent, Belgium.
| |
Collapse
|
8
|
Alshalalfa M, Goglia AG, Swami N, Nguyen B, Hougen HY, Khan A, Kishan AU, Punnen S, Nguyen PL, Mahal BA, Dee EC. Determinants of widespread metastases and of metastatic tropism in patients with prostate cancer: A genomic analysis of primary and metastatic tumors. Urol Oncol 2023; 41:253.e21-253.e26. [PMID: 37003878 PMCID: PMC10559314 DOI: 10.1016/j.urolonc.2023.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/08/2023] [Accepted: 02/20/2023] [Indexed: 04/03/2023]
Abstract
INTRODUCTION Emerging evidence suggests that metastasis is better described as a spectrum of disease rather than a binary state. A greater understanding of the genomic features that determine extent and location of metastatic spread may inform risk stratification and monitoring. Here, we identify genomic alterations from primary prostate carcinomas that are predictive of wide-spread metastatic potential. METHODS Genomic and clinical data from 1,312 patients with primary prostate carcinoma were extracted from the MSK-MET cohort through cBioPortal. Metastatic site counts and overall survival (OS) data were publicly available and used as the primary outcomes. Primary tumor samples were profiled using the MSK-IMPACT targeted sequencing platform. We focused on 58 genes frequently altered in prostate cancer. Cox proportional hazard analyses defined hazard ratios (HRs) and 95% confidence intervals (CIs) for overall mortality in patients with different metastatic outcomes. RESULTS Out of the 1,312 patients in our cohort, 939 (71%) developed metastases, of whom 113 (8.6%) had metastases to 5 or more distinct anatomical sites (defining wide-spread metastases, WSM). Bone was the most common site of metastasis (36%), and 80% of patients with liver metastases had 4 or more additional sites of metastasis. Among patients with metastasis, increasing number of metastatic sites was associated with increased risk of death (HR: 1.8, 95%CI: 1.63-1.99, P < 0.001). Alterations in the following genes were enriched in tumors from patients with WSM vs. others: TP53 (40% vs. 20%, P < 0.0001), FOXA1-amplification (8% vs. 3%, P = 0.02), AR-amplification (4.4% vs. 1%, P = 0.01), RB1-deletion (5.3% vs. 0.7%, P = 0.001), and BRCA2-deletion (4.4% vs. 0.7%, P = 0.01). Univariable survival analysis showed all these alterations were predictive of OS (P < 0.05). On multivariable analysis, only TP53 mutations, and FOXA1 and AR amplifications were independent prognostic factors. FOXA1 (n = 37) and AR (n = 13) amplifications were mutually exclusive and patients with these experienced very poor OS (HR: 3.57, 95%CI:2.26-5.6, P < 0.001]. CONCLUSIONS We identified genomic alterations (TP53 mutations, FOXA1/AR amplification, RB1/BRCA2 deletion) from primary prostate carcinomas that are predictive of wide-spread metastases and poor outcome.
Collapse
Affiliation(s)
- Mohammed Alshalalfa
- University of Miami Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Alexander G Goglia
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nishwant Swami
- Harvard T.H. Chan School of Public Health and University of Massachusetts Chan Medical School, Worchester, MA, USA
| | - Bastien Nguyen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Helen Y Hougen
- Department of Urology, University of Miller School of Medicine/Desai Sethi Urology Institute, Miami, FL, USA
| | - Anwar Khan
- University of Miami Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Amar U Kishan
- Department of Radiation Oncology, David Geffen School of Medicine UCLA/Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Sanoj Punnen
- Department of Urology, University of Miller School of Medicine/Desai Sethi Urology Institute, Miami, FL, USA
| | - Paul L Nguyen
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Harvard Cancer Center, Boston, MA, USA
| | - Brandon A Mahal
- Department of Radiation Oncology, University of Miami Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL, USA.
| | - Edward Christopher Dee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
9
|
Yekedüz E, McKay RR, Gillessen S, Choueiri TK, Ürün Y. Visceral Metastasis Predicts Response to New Hormonal Agents in Metastatic Castration-Sensitive Prostate Cancer. Oncologist 2023:7135855. [PMID: 37084289 DOI: 10.1093/oncolo/oyad102] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/22/2023] [Indexed: 04/23/2023] Open
Abstract
Visceral metastasis (VM) and a higher number of bone metastasis generally define high volume/risk in patients with metastatic castration-sensitive prostate cancer (mCSPC). Subgroup analysis of pivotal trials did not show a clear benefit of second-generation non-steroidal anti-androgens (NSAAs) in patients with VM. However, subgroup analysis of the trial assessing abiraterone acetate, a CYP 17 inhibitor, plus prednisone (AAP) showed an improved overall survival (OS) in patients with mCSPC with VM. We searched MEDLINE, Web of Science, and congress abstracts for the phase III randomized controlled trials of second-generation NSAAs and AAP in patients with mCSPC. In this pooled analysis, we included 6485 patients from the 6 phase III trials. The rate of patients with VM was 15.2%. Interestingly, in contrast to NSAAs, AAP seems to be effective in improving OS among patients with VM (hazard ratio, HR: 0.89, 95% CI, 0.72-1.11, P = .30 for second-generation NSAAs; HR: 0.58, 95% CI, 0.40-0.84, P = .004 for AAP). In contrast, both second-generation NSAAs (HR: 0.63, 95% CI, 0.57-0.70, P < .001) and AAP (HR: 0.68, 95% CI, 0.57-0.81, P < .001) improved OS in patients without VM. In this pooled analysis, we demonstrate that while AAP provided an OS improvement in patients with VM, second-generation NSAAs did not demonstrate a similar OS benefit in this population.
Collapse
Affiliation(s)
- Emre Yekedüz
- Medical Oncology Clinic, Ankara Etlik City Hospital, Ankara, Türkiye
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
- Department of Medical Oncology, Università della Svizzera Italiana, Lugano, Switzerland
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
10
|
Sutera P, Deek MP, Van der Eecken K, Shetty AC, Chang JH, Hodges T, Song Y, Verbeke S, Van Dorpe J, Fonteyne V, De Laere B, Mishra M, Rana Z, Molitoris J, Ferris M, Ross A, Schaeffer E, Roberts N, Song DY, DeWeese T, Pienta KJ, Antonarakis ES, Ost P, Tran PT. WNT Pathway Mutations in Metachronous Oligometastatic Castration-Sensitive Prostate Cancer. Int J Radiat Oncol Biol Phys 2023; 115:1095-1101. [PMID: 36708787 PMCID: PMC10443895 DOI: 10.1016/j.ijrobp.2022.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 01/28/2023]
Abstract
PURPOSE WNT signaling is a cellular pathway that has been implicated in the development and progression of prostate cancer. Oligometastatic castration-sensitive prostate cancer (omCSPC) represents a unique state of disease in which metastasis-directed therapy (MDT) has demonstrated improvement in progression-free survival. Herein, we investigate the clinical implications of genomic alterations in the WNT signaling cascade in men with omCSPC. METHODS AND MATERIALS We performed an international multi-institutional retrospective study of 277 men with metachronous omCSPC who underwent targeted DNA sequencing of their primary/metastatic tumor. Patients were classified by presence or absence of pathogenic WNT pathway mutations (in the genes APC, RNF43, and CTNNB1). Pearson χ2 and Mann-Whitney U tests were used to determine differences in clinical factors between genomic strata. Kaplan-Meier survival curves were generated for radiographic progression-free survival and overall survival, stratified according to WNT pathway mutation status. RESULTS A pathogenic WNT pathway mutation was detected in 11.2% of patients. Patients with WNT pathway mutations were more likely to have visceral metastases (22.6% vs 2.8%; P < .01) and less likely to have regional lymph node metastases (29.0% vs 50.4%; P = .02). At time of oligometastasis, these patients were treated with MDT alone (33.9%), MDT + limited course of systemic therapy (20.6%), systemic therapy alone (22.4%), or observation (defined as no treatment for ≥6 months after metastatic diagnosis). Multivariable cox regression demonstrated WNT pathway mutations associated with significantly worse overall survival (hazard ratio, 3.87; 95% confidence interval, 1.25-12.00). CONCLUSIONS Somatic WNT pathway alterations are present in approximately 11% of patients with omCSPC and are associated with an increased likelihood of visceral metastases. Although these patients have a worse natural history, they may benefit from MDT.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Matthew P Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Kim Van der Eecken
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jin Hee Chang
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Theresa Hodges
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland; Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sofie Verbeke
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Valérie Fonteyne
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Bram De Laere
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Department of Medical Epidemiology, Biostatistics Karolinska Institute, Stockholm, Sweden
| | - Mark Mishra
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Zaker Rana
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jason Molitoris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew Ferris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashley Ross
- Department of Urology, Northwestern University, Chicago, Illinois
| | - Edward Schaeffer
- Department of Urology, Northwestern University, Chicago, Illinois
| | - Nicholas Roberts
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Daniel Y Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; James Buchanan Brady Urologic Institute, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Theodore DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; James Buchanan Brady Urologic Institute, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kenneth J Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; James Buchanan Brady Urologic Institute, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Emmanuel S Antonarakis
- Department of Medicine, University of Minnesota School of Medicine, Minneapolis, Minnesota
| | - Piet Ost
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Department of Radiation Oncology, Iridium Network, Antwerp, Belgium.
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
11
|
Oing C, Bristow RG. Systemic treatment of metastatic hormone-sensitive prostate cancer-upfront triplet versus doublet combination therapy. ESMO Open 2023; 8:101194. [PMID: 36947986 PMCID: PMC10040503 DOI: 10.1016/j.esmoop.2023.101194] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 03/24/2023] Open
Affiliation(s)
- C Oing
- Translational and Clinical Research Institute, Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Sir Bobby Robson Cancer Trials Research Centre, Northern Centre for Cancer Care, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - R G Bristow
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
12
|
Prostate and metastasis diffusion volume based on apparent diffusion coefficient as a prognostic factor in Hormone-naïve prostate Cancer. Clin Exp Metastasis 2023; 40:187-195. [PMID: 36914924 DOI: 10.1007/s10585-023-10200-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
In this study, to assess the utility of whole-body DWI (WB-DWI) as an imaging biomarker for metastatic hormone-naïve prostate cancer (mHNPC), we evaluated tumor diffusion volume based on apparent diffusion coefficient (ADC) values. WB-DWI results obtained from 62 mHNPC patients were evaluated in this retrospective analysis. The association with castration resistant-free survival (CFS) was evaluated for both prostate and metastatic tumor diffusion volume (pDV and mDV, respectively) based on WB-DWI. The usefulness of pDV and mDV based on ADC values to predict CFS was also examined. During the follow-up period, 22 patients progressed to castration-resistant prostate cancer, and the median CFS was 42.6 months. The median mDV and pDV were 6.7 and 12.6 mL, respectively. mDV was a significant predictor of CFS (hazard ratio [HR]: 2.75; p = 0.022), while pDV was not significant. When DV was divided into groups by ADC values (× 10- 3 mm2/s) of 0.4-1.0 and 1.0-1.8 (× 10- 3 mm2/s), mDV with ADC values (× 10- 3 mm2/s) of 0.4-1.0 (mDV0.4-1.0) showed a more favorable association with CFS compared to total mDV. On multivariate analysis, mDV0.4-1.0 and Gleason grade group had a statistically significant association with CFS (HR: 4.0; p = 0.004, and HR: 3.4; p = 0.006, respectively), while pDV with ADC values (× 10- 3 mm2/s) of 0.4-1.0 did not have a significant association. mDV is useful for predicting CFS in mHNPC patients. mDV may be a better imaging biomarker when based on ADC values.
Collapse
|
13
|
Ma TM, Czernin J, Felix C, Alano R, Wilhalme H, Valle L, Steinberg ML, Dahlbom M, Reiter RE, Rettig MB, Cao M, Calais J, Kishan AU. LUNAR: a randomized Phase 2 study of 177 Lutetium-PSMA Neoadjuvant to Ablative Radiotherapy for Oligorecurrent Prostate Cancer (clinical trial protocol). BJU Int 2023. [PMID: 36797449 DOI: 10.1111/bju.15988] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
OBJECTIVE To assess the efficacy of 177 Lu-PNT2002, a novel radiolabelled small molecule that binds with high affinity to prostate-specific membrane antigen (PSMA), in combination with stereotactic body radiotherapy (SBRT) to all sites of metastasis, vs SBRT alone, in men with oligorecurrent metastatic hormone-sensitive prostate cancer (mHSPC). PATIENTS AND METHODS The 177 Lutetium-PSMA Neoadjuvant to Ablative Radiotherapy for Oligorecurrent Prostate Cancer (LUNAR) trial is an open-label, randomized, stratified, two-arm, single-centre, Phase 2 trial to compare the efficacy and safety of neoadjuvant 177 Lu-PNT2002 plus SBRT vs SBRT alone in men with oligorecurrent mHSPC. Key eligibility criteria include one to five lesions identified on a PSMA positron emission tomography (PET)/computed tomography (CT) scan centrally reviewed by a board-certified nuclear medicine physician. Key exclusion criteria include castrate-resistant disease, de novo oligometastatic disease and receipt of androgen deprivation therapy (ADT) within 6 months of trial enrolment. The trial aims to enrol 100 patients who will be centrally randomized to one of the two treatment arms, in a 1:1 ratio. Patients in the control arm receive SBRT to all sites of disease. Patients in the experimental arm receive two cycles of neoadjuvant 177 Lu-PNT2002 (6.8 GBq) 6-8 weeks apart, followed by an interval PSMA PET/CT in 4-6 weeks and dose-adapted SBRT to all sites of disease 1-2 weeks later. The primary endpoint is progression-free survival. Secondary endpoints are radiographic and prostate-specific antigen-based progression, acute and late physician-scored toxicity, patient-reported quality of life, ADT-free survival, time to progression, overall survival, locoregional control, and duration of response. Enrolment in the study commenced in September 2022. RESULTS AND CONCLUSIONS The addition of 177 Lu-PNT2002 to metastasis-directed therapy alone may potentially further forestall disease progression. The results of this Phase 2 trial will determine, for the first time in a randomized fashion, the added benefit of 177 Lu-PNT2002 to SBRT in patients with oligorecurrent mHSPC.
Collapse
Affiliation(s)
- Ting Martin Ma
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA.,Physics and Biology in Medicine Interdepartmental Graduate Program, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Carol Felix
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| | - Rejah Alano
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Holly Wilhalme
- Department of Medicine Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Luca Valle
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| | - Michael L Steinberg
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| | - Magnus Dahlbom
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA.,Physics and Biology in Medicine Interdepartmental Graduate Program, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Robert E Reiter
- Department of Urology, University of California, Los Angeles, CA, USA
| | - Matthew B Rettig
- Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Minsong Cao
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA.,Physics and Biology in Medicine Interdepartmental Graduate Program, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA.,Department of Urology, University of California, Los Angeles, CA, USA
| |
Collapse
|
14
|
Krausewitz P, Ritter M, Essler M. [Diagnosis and staging of metastatic hormone-sensitive prostate cancer]. UROLOGIE (HEIDELBERG, GERMANY) 2023; 62:347-353. [PMID: 36735034 DOI: 10.1007/s00120-023-02028-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Risk stratification of patients with metastatic hormone-sensitive prostate cancer (mHSPC) has undergone significant changes in recent years in light of new therapies and innovative imaging. OBJECTIVES Established and innovative methods for detection of metastasis, risk group stratification, and treatment of mHSPC are outlined and compared. MATERIALS AND METHODS Background knowledge and treatment-relevant guideline recommendations on mHSPC are presented and complemented by recent study results. RESULTS Integration of modern imaging techniques, especially prostate-specific membrane antigen (PSMA) PET/CT, into the diagnostic algorithm has the potential to significantly improve risk stratification and treatment of mHSPC. By using PSMA PET/CT, metastases are detected early and sensitively. This leads to the definition of new subgroups amenable to modern therapeutic strategies. The prognostic value of using PSMA PET/CT with regard to established risk categories in mHSPC is currently being evaluated. CONCLUSIONS Modern imaging, especially PSMA PET/CT, has significant added value for the diagnosis and treatment of mHSPC in almost all subgroups. In particular, it helps to select patients who will benefit from intensification or de-escalation of systemic therapy.
Collapse
Affiliation(s)
- Philipp Krausewitz
- Klinik und Poliklinik für Urologie und Kinderurologie, Universitätsklinikum Bonn (AöR), Venusberg-Campus 1, 53127, Bonn, Deutschland.
| | - Manuel Ritter
- Klinik und Poliklinik für Urologie und Kinderurologie, Universitätsklinikum Bonn (AöR), Venusberg-Campus 1, 53127, Bonn, Deutschland
| | - Markus Essler
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Bonn (AöR), Venusberg-Campus 1, 53127, Bonn, Deutschland
| |
Collapse
|
15
|
Tran PT, Sutera P, Phillips RM, Deek MP, Chmura S. From Idea to Clinical Practice: A Brief History of Oligometastatic Disease. Int J Radiat Oncol Biol Phys 2022; 114:576-580. [DOI: 10.1016/j.ijrobp.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/03/2022] [Indexed: 10/31/2022]
|
16
|
Sutera P, Deek MP, Van der Eecken K, Wyatt AW, Kishan AU, Molitoris JK, Ferris MJ, Minhaj Siddiqui M, Rana Z, Mishra MV, Kwok Y, Davicioni E, Spratt DE, Ost P, Feng FY, Tran PT. Genomic biomarkers to guide precision radiotherapy in prostate cancer. Prostate 2022; 82 Suppl 1:S73-S85. [PMID: 35657158 PMCID: PMC9202472 DOI: 10.1002/pros.24373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/30/2022] [Accepted: 04/29/2022] [Indexed: 11/08/2022]
Abstract
Our ability to prognosticate the clinical course of patients with cancer has historically been limited to clinical, histopathological, and radiographic features. It has long been clear however, that these data alone do not adequately capture the heterogeneity and breadth of disease trajectories experienced by patients. The advent of efficient genomic sequencing has led to a revolution in cancer care as we try to understand and personalize treatment specific to patient clinico-genomic phenotypes. Within prostate cancer, emerging evidence suggests that tumor genomics (e.g., DNA, RNA, and epigenetics) can be utilized to inform clinical decision making. In addition to providing discriminatory information about prognosis, it is likely tumor genomics also hold a key in predicting response to oncologic therapies which could be used to further tailor treatment recommendations. Herein we review select literature surrounding the use of tumor genomics within the management of prostate cancer, specifically leaning toward analytically validated and clinically tested genomic biomarkers utilized in radiotherapy and/or adjunctive therapies given with radiotherapy.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew P. Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Kim Van der Eecken
- Department of Pathology, Ghent University Hospital, Cancer Research Institute (CRIG), Ghent, Belgium
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amar U. Kishan
- Department of Radiation Oncology, UCLA, Los Angeles, CA, USA
| | - Jason K. Molitoris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew J. Ferris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M. Minhaj Siddiqui
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zaker Rana
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark V. Mishra
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Young Kwok
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Daniel E. Spratt
- Department of Radiation Oncology, University Hospitals, Cleveland, OH, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, Antwerp, Belgium and Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Felix Y. Feng
- Departments of Radiation Oncology, Medicine and Urology, UCSF, San Francisco, CA, USA
| | - Phuoc T. Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
The oligometastatic spectrum in the era of improved detection and modern systemic therapy. Nat Rev Clin Oncol 2022; 19:585-599. [PMID: 35831494 DOI: 10.1038/s41571-022-00655-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
Metastases remain the leading cause of cancer-related mortality. The oligometastasis hypothesis postulates that a spectrum of metastatic spread exists and that some patients with a limited burden of metastases can be cured with ablative therapy. Over the past decade, substantial advances in systemic therapies have resulted in considerable improvements in the outcomes of patients with metastatic cancers, warranting re-examination of the oligometastatic paradigm and the role of local ablative therapies within the context of the improved therapeutic responses, shifting patterns of disease recurrence and possible synergy with systemic treatments. Herein, we reframe the oligometastatic phenotype as a dynamic state for which locally ablative, metastasis-directed therapy improves clinical outcomes, including by prolonging survival and increasing cure rates. Important risk factors defining the metastatic spectrum are highlighted that inform both staging and therapy. Finally, we synthesize the literature on combining local therapies with modern systemic treatments, identifying general themes to optimally integrate ablative therapies in this context.
Collapse
|