1
|
Rahman R, Selth LA. Cyclin-dependent kinases as mediators of aberrant transcription in prostate cancer. Transl Oncol 2025; 55:102378. [PMID: 40163908 PMCID: PMC11995790 DOI: 10.1016/j.tranon.2025.102378] [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: 11/24/2024] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Transcriptional control of gene expression is fundamental to all cellular processes. Conversely, transcriptional dysregulation is a hallmark of cancer. While this hallmark is a key driver of all malignancy-related process, it also represents a vulnerability that can be exploited therapeutically. Prostate cancer is a prime example of this phenomenon: it is characterised by aberrant transcription and treated with drugs that influence transcriptional pathways. Indeed, the primary oncogenic driver and therapeutic target of prostate cancer, the androgen receptor (AR), is a transcription factor. Moreover, a plethora of other transcriptional regulators, including transcriptional cyclin-dependent kinases (CDK7, CDK8 and CDK9), MYC and Bromodomain-containing protein 4 (BRD4), play prominent roles in disease progression. In this review, we focus on the roles of transcriptional CDKs in prostate cancer growth, metastasis and therapy resistance and discuss their interplay with AR, MYC and BRD4. Additionally, we explore recent advances in the therapeutic targeting of transcriptional CDKs and propose how these strategies could be effectively harnessed for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Razia Rahman
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia
| | - Luke A Selth
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia; Flinders University, Freemasons Centre for Male Health and Wellbeing, Adelaide, South Australia; Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
2
|
Freitas PFS, Abdshah A, McKay RR, Sharifi N. HSD3B1, prostate cancer mortality and modifiable outcomes. Nat Rev Urol 2025; 22:313-320. [PMID: 39543357 DOI: 10.1038/s41585-024-00953-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2024] [Indexed: 11/17/2024]
Abstract
Androgen receptor stimulation by testosterone and dihydrotestosterone is crucial for prostate cancer progression. Despite the initial effectiveness of androgen deprivation therapy (ADT), castration-resistant prostate cancer eventually develops in most men. A common germline missense-encoding polymorphism in HSD3B1 increases extra-gonadal androgen biosynthesis from adrenal precursors owing to increased availability of the encoded enzyme 3β-hydroxysteroid dehydrogenase 1 (3βHSD1) - hence, it is called the adrenal-permissive enzyme. This mechanism explains the more rapid progression to castration-resistant prostate cancer in men who inherit this allele than in men without it via sustained androgen receptor activation despite ADT. Multiple clinical studies, including data derived from prospective phase III studies, have linked adrenal-permissive allele inheritance to inferior clinical responses to ADT and increased mortality, but reversal is possible with upfront adrenal androgen blockade. The adrenal-permissive allele exhibits divergent frequencies across various groups worldwide, which could contribute to differences in clinical outcomes among these populations. Large-scale data from the Million Veteran Program have shown homozygous HSD3B1 adrenal-permissive allele inheritance to be an independent biomarker of prostate cancer-specific mortality. Together, these observations support the integration of HSD3B1 into germline testing and clinical trials as it might help to identify groups at increased likelihood of benefiting from early, intensified, AR-targeting interventions. Lastly, 3βHSD1 is a promising target for pharmacological inhibition, which enables new strategies for systemic prostate cancer therapy.
Collapse
Affiliation(s)
- Pedro F S Freitas
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alireza Abdshah
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rana R McKay
- Division of Hematology-Oncology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nima Sharifi
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
3
|
Yu EY, Rumble RB, Agarwal N, Cheng HH, Eggener SE, Bitting RL, Beltran H, Giri VN, Spratt D, Mahal B, Lu K, Crispino T, Trabulsi EJ. Germline and Somatic Genomic Testing for Metastatic Prostate Cancer: ASCO Guideline. J Clin Oncol 2025; 43:748-758. [PMID: 39787437 DOI: 10.1200/jco-24-02608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE To evaluate evidence on germline and somatic genomic testing for patients with metastatic prostate cancer and provide recommendations. METHODS A systematic review by a multidisciplinary panel with patient representation was conducted. The PubMed database was searched from January 2018 to May 2024. Articles were selected for inclusion if they reported on patients with metastatic prostate cancer who received a germline or somatic genomic test and/or made comparisons between those tests, reported detection rates, prognostic information, or treatment implications. RESULTS A total of 1,713 papers were identified in the literature search. After applying the eligibility criteria, 14 remained: eight systematic reviews and six clinical trials. RECOMMENDATIONS Patients with metastatic prostate cancer should undergo both germline and somatic DNA sequencing using panel-based assays. These tests can guide the use of poly(ADP-ribose) polymerase inhibitors, which have a survival benefit in metastatic castration-resistant prostate cancer. In addition, germline testing may have screening implications for additional cancers for patients and cascade testing implications for family members. The data supporting when to perform repeat testing and optimal tissue type to use (eg, primary tumor v metastatic biopsy versus circulating tumor DNA [ctDNA] testing) are more limited, but this panel recommends considering retesting in patients whose results were previously negative or uninformative, and to consider either a metastatic biopsy or ctDNA when a significant change in clinical status occurs. Next-generation genomic sequencing findings that are associated with prognostic only (and not predictive) value should not be used to guide treatment outside of a clinical trial.Additional information is available at www.asco.org/genitourinary-cancer-guidelines.
Collapse
Affiliation(s)
- Evan Y Yu
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Neeraj Agarwal
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT
| | - Heather H Cheng
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | | | - Veda N Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, CT
| | | | | | - Kevin Lu
- Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Tony Crispino
- Patient Representative, Chapter President UsTOO Las Vegas, Prostate Cancer Education and Support, Las Vegas, NV
| | - Edouard J Trabulsi
- Jefferson Einstein Medical Center, Sidney Kimmel Cancer Center of Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
4
|
Gebrael G, Sayegh N, Hage Chehade C, Jo Y, Narang A, Chigarira B, Tripathi N, Srivastava A, Tandar C, Williams JF, Garg D, Ji R, Maughan BL, Swami U, Agarwal N. Genomic biomarkers of survival in patients with metastatic hormone-sensitive prostate cancer undergoing intensified androgen deprivation therapy. Prostate Cancer Prostatic Dis 2025:10.1038/s41391-025-00936-1. [PMID: 39885371 DOI: 10.1038/s41391-025-00936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/04/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
INTRODUCTION Androgen deprivation therapy intensification (ADTi) with androgen receptor pathway inhibitors (ARPI), docetaxel or both has been shown to improve survival outcomes in patients with metastatic hormone-sensitive prostate cancer (mHSPC). Currently, baseline tumor genomic markers have no role in clinical decision-making in patients with mHSPC. METHODS In this IRB-approved retrospective study, patients diagnosed with mHSPC who underwent comprehensive genomic profiling from primary tissue or metastatic sites and treated with ADTi were included. Genomic alterations with an incidence ≥5% were included in the analysis. RESULTS A total of 276 patients were eligible and included in the study. In the multivariable analysis, TP53 (HR 1.71, 95% CI 1.17-2.49, p = 0.006), RB1 (HR 2.32, 95% CI 1.28-4.18, p = 0.006), PTEN (HR 1.74, 95% CI 1.12-2.7, p = 0.014), and BRCA2 (HR 2.64, 95% CI 1.42-4.92, p = 0.003) were associated with significantly shorter PFS, while TP53 (HR 1.63, 95% CI 1.00-2.64, p = 0.049), RB1 (HR 4.5, 95% CI 2.32-8.70, p < 0.001), and PTEN (HR 2.4, 95% CI 1.38-4.2, p = 0.003) were associated with significantly worse OS. CONCLUSIONS This is one of the largest studies to show the association of baseline tumor genomic markers with survival in patients with mHSPC treated with ADTi. Upon external validation, these results may aid in developing a clinical-genomic risk stratification model, patient counseling, and prognostication.
Collapse
Affiliation(s)
- Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicolas Sayegh
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chadi Hage Chehade
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Yeonjung Jo
- Division of Biostatistics, Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Cancer Biostatistics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Arshit Narang
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Beverly Chigarira
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nishita Tripathi
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ayana Srivastava
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Clara Tandar
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Diya Garg
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Richard Ji
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin L Maughan
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
5
|
Guo H, Jin C, Ding L, Xie J, Xu J, Wang R, Wang H, Guo C, Zhang J, Peng B, Yao X, Yuan J, Yang B. De novo patients with high-volume metastatic hormone-sensitive prostate cancer can benefit from the addition of docetaxel to triplet therapy: Network-analysis and systematic review. Chin Med J (Engl) 2025; 138:231-233. [PMID: 39350461 PMCID: PMC11745853 DOI: 10.1097/cm9.0000000000003311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Indexed: 01/21/2025] Open
Affiliation(s)
- Hanxu Guo
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Chengqi Jin
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
| | - Li Ding
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Jun Xie
- Department of Urology, Shanghai Clinical College, Anhui Medical University, Shanghai 200072, China
| | - Jing Xu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Ruiliang Wang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Hong Wang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Changcheng Guo
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiansheng Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Bo Peng
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Shanghai Clinical College, Anhui Medical University, Shanghai 200072, China
- Department of Urology, Shanghai Tenth People’s Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
- Department of Urology, Shanghai Clinical College, Anhui Medical University, Shanghai 200072, China
- Department of Urology, Shanghai Tenth People’s Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jing Yuan
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Bin Yang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
- Department of Urology, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
- Department of Urology, Shanghai Tenth People’s Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
6
|
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; 7:1403-1410. [PMID: 38570239 DOI: 10.1016/j.euo.2024.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
7
|
Wang T, Kim J, Kumar R, Deek RA, Stephenson R, Mayer T, Saraiya B, Ghodoussipour S, Jang T, Golombos D, Packiam V, Ennis R, Hathout L, Jabbour SK, Guler O, Onal C, Deek MP. Landscape and prognostic significance of oncogene drivers in metastatic castration sensitive prostate cancer. Transl Cancer Res 2024; 13:6235-6245. [PMID: 39697761 PMCID: PMC11651787 DOI: 10.21037/tcr-24-123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/22/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Tumor suppressors are well known drivers of cancer invasion and metastasis in metastatic castration sensitive prostate cancer (mCSPC). However, oncogenes are also known to be altered in this state, however the frequency and prognosis of these alterations are unclear. Thus, we aimed to study the spectrum of oncogene mutations in mCSPC and study the significance of these alteration on outcomes. METHODS Four hundred and seventy-seven patients with mCSPC were included who underwent next generation sequencing. Oncogene alterations were defined as mutations in ALK, AKT1-3, BRAF, CCND1-3, CTNNB1, EGFR, ERBB2, FGFR1, FGFR2, HRAS, KRAS, MDM2, MET, MITF, MYC, NOTCH1-3, NRAS, PIK3CA, PI3KCB, PIK3R1, RET. Endpoints of interests were radiographic progression-free survival (rPFS), time to development of CRPC (tdCRPC), and overall survival (OS). Kaplan Meier analysis was performed and Cox regression hazard ratios (HR) calculated. RESULTS A total of 477 patients were included with baseline characteristics with 117 patients (24.5%) harbored a mutation within an oncogene. A total of 172 oncogene mutations were found within the population with the most common being MYC (n=29; 16.9%), PIK3CA (n=24; 14%), CTNNB1 (n=22, 12.8%), BRAF (n=10, 5.8%), and CCND1 (n=10, 5.8%). Oncogene mutations were associated with inferior rPFS (19.2 vs. 32.2 months, P=0.03), tdCRPC (15.7 vs. 32.4 months, P<0.001), and OS (5-year OS 75.3% vs. 55.4%, P=0.01). On multivariable analysis oncogene mutations were strongly associated with tdCRPC (HR 1.42, P=0.03). CONCLUSIONS Oncogenes are frequency mutated in mCSPC and associated with aggressive features and inferior outcomes. Future work will need to validate these results to better assess its significance in allowing for personalization of care.
Collapse
Affiliation(s)
- Theodore Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Jongmyung Kim
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Ritesh Kumar
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Rebecca A. Deek
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan Stephenson
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Tina Mayer
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Biren Saraiya
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Saum Ghodoussipour
- Department of Urology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Thomas Jang
- Department of Urology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - David Golombos
- Department of Urology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Vignesh Packiam
- Department of Urology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Ronald Ennis
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Lara Hathout
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Ozan Guler
- Department of Radiation Oncology, Baskent University, Ankara, Turkey
| | - Cem Onal
- Department of Radiation Oncology, Baskent University, Ankara, Turkey
| | - Matthew P. Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
8
|
Sutera P, Kim J, Kumar R, Deek RA, Stephenson R, Mayer T, Saraiya B, Ghodoussipour S, Jang T, Golombos D, Packiam V, Ennis R, Hathout L, Jabbour SK, Guler O, Onal C, Tran PT, Deek MP. PIK3/Akt/mTOR pathway alterations in metastatic castration-sensitive prostate cancer. Prostate 2024; 84:1301-1308. [PMID: 39021052 DOI: 10.1002/pros.24765] [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: 01/28/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Alterations in the PIK3/Akt/mTOR pathway are commonly seen in metastatic castration-sensitive prostate cancer (mCSPC), however their role in outcomes is unknown. We aim to evaluate the prognostic significance as well as the genetic landscape of PIK3/Akt/mTOR pathway alteration in mCSPC. METHODS Fourhundred and seventy-two patients with mCSPC were included who underwent next generation sequencing. PIK3/Akt/mTor pathway alterations were defined as mutations in Akt1, mTOR, PIK3CA, PIK3CB, PIK3R1, PTEN, TSC1, and TSC2. Endpoints of interests were radiographic progression-free survival (rPFS), time to development of castration resistant prostate cancer (tdCRPC), and overall survival (OS). Kaplan-Meier analysis was performed and Cox regression hazard ratios (HR) were calculated. RESULTS One hundred and fifty-two (31.9%) patients harbored a PIK3/Akt/mTOR pathway alteration. Median rPFS and tdCRPC were 23.7 and 21.0 months in PIK3/Akt/mTOR altered compared to 32.8 (p = 0.08) and 32.1 months (p = 0.002) in wildtype tumors. On multivariable analysis PIK3/Akt/mTOR pathway alterations were associated with tdCRPC (HR 1.43, 95% CI, 1.05-1.94, p = 0.02), but not rPFS [Hazard ratio (HR) 1.20, 95% confidence interval (CI), 0.90-1.60, p = 0.21]. PIK3/Akt/mTOR pathway alterations were more likely to be associated with concurrent mutations in TP53 (40% vs. 28%, p = 0.01) and TMPRSS2-ERG (37% vs. 26%, p = 0.02) than tumors without PIK3/Akt/mTOR pathway alterations. Concurrent mutations were typically associated with shorter median times to rPFS and tdCRPC. DAVID analysis showed p53 signaling and angiogenesis pathways were enriched in PIK3/Akt/mTOR pathway altered tumors while beta-catenin binding and altered BRCA pathway were enriched in PIK3/Akt/mTOR pathway wildtype tumors. CONCLUSIONS PIK3/Akt/mTOR pathway alterations were common in mCSPC and associated with poorer prognosis. The genetic landscape of PIK3/Akt/mTOR pathway altered tumors differed from wildtype tumors. Additional studies are needed to better understand and target the PIK3/Akt/mTOR pathway in mCSPC.
Collapse
Affiliation(s)
- Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jongmyung Kim
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Ritesh Kumar
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Rebecca A Deek
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan Stephenson
- Rutgers Robert Wood Johnson Medical School, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Tina Mayer
- Rutgers Robert Wood Johnson Medical School, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Biren Saraiya
- Rutgers Robert Wood Johnson Medical School, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Saum Ghodoussipour
- Department of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Thomas Jang
- Department of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - David Golombos
- Department of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Vignesh Packiam
- Department of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, USA
| | - Ronald Ennis
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lara Hathout
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Salma K Jabbour
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ozan Guler
- Department of Radiation Oncology, Baskent University, Ankara, Turkey
| | - Cem Onal
- Department of Radiation Oncology, Baskent University, Ankara, Turkey
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew P Deek
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Corres-Mendizabal J, Zacchi F, Martín-Martín N, Mateo J, Carracedo A. Metastatic hormone-naïve prostate cancer: a distinct biological entity. Trends Cancer 2024; 10:825-841. [PMID: 39048488 PMCID: PMC11397905 DOI: 10.1016/j.trecan.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024]
Abstract
Metastatic hormone-naïve prostate cancer (mHNPC) is often the initial form of presentation for metastatic prostate cancer and encompasses a heterogeneous patient population with high inter-patient heterogeneity in prognosis and response to therapy. A more precise treatment of mHNPC, guided by evidence-based biomarkers, remains an unmet medical need. In addition, the limited number of representative laboratory models of mHNPC hampers the translation of basic research into clinical applications. We provide a comprehensive overview of the clinical and biological features that characterize mHNPC, highlight molecular data that could explain the unique prognostic characteristics of mHNPC, and identify key open questions.
Collapse
Affiliation(s)
- Jon Corres-Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Francesca Zacchi
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy; Vall Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital Campus, Barcelona, Spain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain; Translational Prostate Cancer Research Laboratory, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Joaquin Mateo
- Vall Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital Campus, Barcelona, Spain.
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain; Translational Prostate Cancer Research Laboratory, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| |
Collapse
|
10
|
Jiménez N, Garcia de Herreros M, Reig Ò, Marín-Aguilera M, Aversa C, Ferrer-Mileo L, García-Esteve S, Rodríguez-Carunchio L, Trias I, Font A, Rodriguez-Vida A, Climent MÁ, Cros S, Chirivella I, Domènech M, Figols M, Carles J, Suárez C, Herrero Rivera D, González-Billalabeitia E, Cívico C, Sala-González N, Ruiz de Porras V, Ribal MJ, Prat A, Mellado B. Development and Independent Validation of a Prognostic Gene Expression Signature Based on RB1, PTEN, and TP53 in Metastatic Hormone-sensitive Prostate Cancer Patients. Eur Urol Oncol 2024; 7:954-964. [PMID: 38429210 DOI: 10.1016/j.euo.2023.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/29/2023] [Indexed: 03/03/2024]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) with docetaxel (D) and/or antiandrogen receptor therapies (ARTs) are the standard therapies in metastatic hormone-sensitive prostate cancer (mHSPC). Alterations in the tumor suppressor genes (TSGs) RB1, PTEN, and TP53 are associated with an aggressive evolution and treatment resistance in castration-resistant prostate cancer (CRPC). OBJECTIVE To study the clinical implications of TSG mRNA expression in mHSPC patients. DESIGN, SETTING, AND PARTICIPANTS This is a multicenter retrospective biomarker study in mHSPC patients. TSGlow status was defined when two or more out of the three TSGs presented low RNA expression by nCounter in formalin-fixed paraffin-embedded samples and TSGwt for the remaining cases. The microarray data from the CHAARTED trial were analyzed as an independent validation cohort. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Molecular data were correlated with CRPC-free survival (CRPC-FS) and overall survival (OS) by the Kaplan-Meier method and multivariate Cox analysis. RESULTS AND LIMITATIONS A total of 226 patients were included, of whom 218 were eligible: 93 were treated with ADT and 125 with ADT + D; 75.7% presented de novo stage IV and 67.9% high-volume disease. TSGlow (19.2%) was independently correlated with shorter CRPC-FS (hazard ratio [HR] 1.8, p = 0.002) and OS (HR 2, p = 0.002). In the CHAARTED trial, TSGlow was independently correlated with lower CRPC-FS (HR 2.2, p = 0.02); no differences in clinical outcomes according to treatment were observed in TSGlow patients, while a significant benefit was observed for ADT + D in the TSGwt group for CRPC-FS (HR 0.4, p < 0.001) and OS (HR 0.4, p = 0.001). However, no interaction was observed between TSG signature and treatment in either series. Study limitations are the retrospective design, small sample size, and lack of inclusion of patients treated with ADT + ART. CONCLUSIONS TSGlow expression correlates with adverse outcomes in patients with mHSPC. The investigation of new therapeutic strategies in these patients is warranted. PATIENT SUMMARY The low RNA expression of tumor suppressor genes in the tumors is correlated with adverse outcomes in patients with metastatic hormone-sensitive prostate cancer.
Collapse
Affiliation(s)
- Natalia Jiménez
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Marta Garcia de Herreros
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clínic, Barcelona, Spain
| | - Òscar Reig
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clínic, Barcelona, Spain; Uro-Oncology Unit, Hospital Clínic, University of Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Mercedes Marín-Aguilera
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clínic, Barcelona, Spain
| | - Caterina Aversa
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clínic, Barcelona, Spain; Uro-Oncology Unit, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Laura Ferrer-Mileo
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clínic, Barcelona, Spain; Uro-Oncology Unit, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Samuel García-Esteve
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Leonardo Rodríguez-Carunchio
- Uro-Oncology Unit, Hospital Clínic, University of Barcelona, Barcelona, Spain; Department of Pathology, Hospital Clínic, Barcelona, Spain
| | - Isabel Trias
- Uro-Oncology Unit, Hospital Clínic, University of Barcelona, Barcelona, Spain; Department of Pathology, Hospital Clínic, Barcelona, Spain
| | - Albert Font
- Medical Oncology Department, Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Alejo Rodriguez-Vida
- Medical Oncology Department, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Hospital del Mar, Barcelona, Spain
| | - Miguel Ángel Climent
- Medical Oncology Service, Instituto Valenciano de Oncología (IVO), Valencia, Spain
| | - Sara Cros
- Medical Oncology Department, Hospital General de Granollers, Barcelona, Spain
| | - Isabel Chirivella
- Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Montserrat Domènech
- Medical Oncology Department, Fundació Althaia, Xarxa Assistencial Universitària de Manresa, Spain
| | - Mariona Figols
- Medical Oncology Department, Fundació Althaia, Xarxa Assistencial Universitària de Manresa, Spain
| | - Joan Carles
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Cristina Suárez
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | - Claudia Cívico
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, IMIB-Universidad de Murcia, Murcia, Spain
| | | | - Vicenç Ruiz de Porras
- Badalona Applied Research Group in Oncology (B-ARGO), Institut Català d'Oncologia - Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Maria J Ribal
- Department of Urology, Hospital Clínic, Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clínic, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Begoña Mellado
- Translational Genomics and Targeted Therapeutics in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clínic, Barcelona, Spain; Uro-Oncology Unit, Hospital Clínic, University of Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
11
|
Gómez-Aparicio MA, López-Campos F, Buchser D, Lazo A, Willisch P, Ocanto A, Sargos P, Shelan M, Couñago F. Is There an Opportunity to De-Escalate Treatments in Selected Patients with Metastatic Hormone-Sensitive Prostate Cancer? Cancers (Basel) 2024; 16:2331. [PMID: 39001393 PMCID: PMC11240449 DOI: 10.3390/cancers16132331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The treatment landscape for metastatic hormone-sensitive prostate cancer continues to evolve, with systemic treatment being the mainstay of current treatment. Prognostic and predictive factors such as tumour volume and disease presentation have been studied to assess responses to different treatments. Intensification and de-escalation strategies arouse great interest, so several trials are being developed to further personalize the therapy in these populations. Is there an optimal sequence and a possible option to de-intensify treatment in selected patients with a favourable profile? This and other goals will be the subject of this review.
Collapse
Affiliation(s)
| | - Fernando López-Campos
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Department of Radiation Oncology, Hospital Universitario San Francisco de Asis and Hospital Vithas La Milagrosa, GenesisCare, 28002 Madrid, Spain; (A.O.); (F.C.)
| | - David Buchser
- Department of Radiation Oncology, Hospital Universitario Cruces, 48903 Barakaldo, Spain;
| | - Antonio Lazo
- Department of Radiation Oncology, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain;
| | - Patricia Willisch
- Department of Radiation Oncology, Hospital Meixoeiro, 36214 Vigo, Spain;
| | - Abrahams Ocanto
- Department of Radiation Oncology, Hospital Universitario San Francisco de Asis and Hospital Vithas La Milagrosa, GenesisCare, 28002 Madrid, Spain; (A.O.); (F.C.)
| | - Paul Sargos
- Department of Radiation Oncology, Institut Bergonié, 33000 Bordeaux, France;
| | - Mohamed Shelan
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Switzerland;
| | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario San Francisco de Asis and Hospital Vithas La Milagrosa, GenesisCare, 28002 Madrid, Spain; (A.O.); (F.C.)
| |
Collapse
|
12
|
Jian J, Wang X, Zhang J, Zhou C, Hou X, Huang Y, Hou J, Lin Y, Wei X. Molecular landscape for risk prediction and personalized therapeutics of castration-resistant prostate cancer: at a glance. Front Endocrinol (Lausanne) 2024; 15:1360430. [PMID: 38887275 PMCID: PMC11180744 DOI: 10.3389/fendo.2024.1360430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Prostate cancer (PCa) is commonly occurred with high incidence in men worldwide, and many patients will be eventually suffered from the dilemma of castration-resistance with the time of disease progression. Castration-resistant PCa (CRPC) is an advanced subtype of PCa with heterogeneous carcinogenesis, resulting in poor prognosis and difficulties in therapy. Currently, disorders in androgen receptor (AR)-related signaling are widely acknowledged as the leading cause of CRPC development, and some non-AR-based strategies are also proposed for CRPC clinical analyses. The initiation of CRPC is a consequence of abnormal interaction and regulation among molecules and pathways at multi-biological levels. In this study, CRPC-associated genes, RNAs, proteins, and metabolites were manually collected and integrated by a comprehensive literature review, and they were functionally classified and compared based on the role during CRPC evolution, i.e., drivers, suppressors, and biomarkers, etc. Finally, translational perspectives for data-driven and artificial intelligence-powered CRPC systems biology analysis were discussed to highlight the significance of novel molecule-based approaches for CRPC precision medicine and holistic healthcare.
Collapse
Affiliation(s)
- Jingang Jian
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin’an Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaorui Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
13
|
Grisay G, Lavaud P, Fizazi K. Current Systemic Therapy in Men with Metastatic Castration-Sensitive Prostate Cancer. Curr Oncol Rep 2024; 26:488-495. [PMID: 38592590 DOI: 10.1007/s11912-024-01509-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2024] [Indexed: 04/10/2024]
Abstract
PURPOSE OF REVIEW This review aims to explore the evolving landscape of treatments available for metastatic castration-sensitive prostate cancer (mCSPC) patients. RECENT FINDINGS In less than a decade, evidence was chronologically provided that (1) systemic treatment intensification with docetaxel improves outcomes, including survival, in men with mCSPC, (2) then that these outcomes are also improved when a second-generation androgen receptor pathway inhibitor (ARPI) is combined with androgen deprivation therapy (ADT), and (3) using a "triplet systemic therapy," which consists in the combination of ADT, an ARPI and docetaxel, further improves outcomes, including survival. Radiotherapy to the prostate combined with ADT alone is now recommended in men with low-volume mCSPC. Combining prostate radiotherapy and intensified systemic treatment including abiraterone may be synergistic as suggested in the PEACE-1 trial. Also, the role of metastases-directed local therapies (mostly stereotactic radiotherapy) is currently being assessed in phase 3 trials. Finally, the integration of biomarkers (e.g. BRCA2 gene alterations, PTEN loss, PSMA expression) for decision making is not currently established, though trials are also currently underway. Importantly, most evidence currently available was obtained in men with de novo metastases, while for those with metastatic relapse after definitive local treatment, the role of treatment intensification is less well established. Treatment intensification is nowadays the standard of care for patients with de novo mCSPC as it leads to outcomes improvement, including survival, and the standard of care is evolving almost on a yearly basis.
Collapse
Affiliation(s)
- Guillaume Grisay
- Department of Medical Oncology, Centres Hospitaliers Universitaires Helora, La Louvière, Belgium.
| | - Pernelle Lavaud
- Department of Cancer Medicine, Institut Gustave Roussy, University of Paris-Saclay, Villejuif, France
| | - Karim Fizazi
- Department of Cancer Medicine, Institut Gustave Roussy, University of Paris-Saclay, Villejuif, France
| |
Collapse
|
14
|
Menezes RDS, Dornas MC, Campos CFF, Rodeiro DB, Carrerette FB, Oliveira RV, de Souza BA, Alves de Souza Carvalho G, Brito IADA, Silva DA, Damião R, Porto LC. Evaluation of HNF1B, KLK3, ELAC2, TMPRSS2-ERG, and CTNNB1 polymorphisms associated with prostate cancer in samples of patients from HUPE-UERJ. Prostate 2024; 84:166-176. [PMID: 37839045 DOI: 10.1002/pros.24635] [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: 04/28/2023] [Revised: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023]
Abstract
PURPOSE Prostate cancer (PCa) is the leading cause of death among men in 48 countries. Genetic alterations play a significant role in PCa carcinogenesis. For the hypothesis of this research, five unique polymorphisms (SNP) were investigated in different genes that showed to be associated in different ways with PCa: rs4430796, rs2735839, rs4792311, rs12329760, and rs28931588, respectively for the genes HNF1B, KLK3, ELAC2, TMPRSS2-ERG, and CTNNB1. PATIENTS AND METHODS Blood samples from 426 subjects were evaluated: 290 controls (161 females and 129 males) and 136 PCa patients. SNP were determined by real-time polymerase chain reaction. TaqMan SNP genotyping assay. In the control samples, the SNPs were defined in association with the self-reported ethnicity, and in 218 control samples with markers with ancestry indicators. The genes were in Hardy-Weinberg equilibrium. One hundred and seventy control samples were matched by ethnicity for comparison with the PCa samples. RESULTS The G allele at rs28931588 was monomorphic in both patients and controls studied. Significant differences were observed in allelic and genotypic frequencies between the control and Pca samples in rs2735839 (KLK3; p = 0.002 and χ2 = 8.73 and p = 0.01, respectively), by the global frequency and in the dominant model rs2735839_GG (odds ratio [OR] = 0.51, p = 0.02). AA and GA genotypes at rs4792311 (ELAC2) were more frequent in patients with Gleason 7(4 + 3), 8, and 9 (n = 37%-59.7%) compared to patients with Gleason 6 and 7(3 + 4) (n = 26%-40.0%) conferring a protective effect on the GG genotype (OR = 0.45, p = 0.02). The same genotype showed an OR = 2.71 (p = 0.01) for patients with low severity. The HNF1B-KLK3-ELAC2-TMPRSS2-ERG haplotypes: GAAT, AAAT, GAGT, and AAGT were more frequent in patients with Pca with OR ranging from 4.65 to 2.48. CONCLUSIONS Higher frequencies of risk alleles were confirmed in the SNPs, KLK3 rs2735839_A, ELAC2 rs4792311_A, and TMPRSS2 rs12329760_T in patients with Pca. Rs2735839_A was associated with risk of Pca and rs4792311_A with severity and Gleason score of 7(4 + 3) or greater. There is a need for careful observation of rs2735839 and rs4792311 in association with the prostatic biopsy due to the increased risk of Pca.
Collapse
Affiliation(s)
- Raphaela Dos Santos Menezes
- Human and Experimental Biology Graduate Program, IBRAG, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Maria Cristina Dornas
- Urology Teaching Assistance Unit (UDA), FCM, Pedro Ernesto University Hospital (HUPE) and the Piquet Carneiro University Polyclinic (PPC), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carlos Frederico Ferreira Campos
- Anatomopathological Service Pedro Ernesto University Hospital (HUPE), FCM, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Daniela Bouzas Rodeiro
- Urology Teaching Assistance Unit (UDA), FCM, Pedro Ernesto University Hospital (HUPE) and the Piquet Carneiro University Polyclinic (PPC), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Fabricio Borges Carrerette
- Urology Teaching Assistance Unit (UDA), FCM, Pedro Ernesto University Hospital (HUPE) and the Piquet Carneiro University Polyclinic (PPC), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Romulo Vianna Oliveira
- Tissue Repair and Histocompatibility Technological Core (Tixus), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Brenda Amaral de Souza
- Tissue Repair and Histocompatibility Technological Core (Tixus), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | | | - Dayse Aparecida Silva
- Laboratory of DNA Diagnostic, IBRAG, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ronaldo Damião
- Urology Teaching Assistance Unit (UDA), FCM, Pedro Ernesto University Hospital (HUPE) and the Piquet Carneiro University Polyclinic (PPC), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Luís Cristóvão Porto
- Tissue Repair and Histocompatibility Technological Core (Tixus), Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Sood A, Kishan AU, Evans CP, Feng FY, Morgan TM, Murphy DG, Padhani AR, Pinto P, Van der Poel HG, Tilki D, Briganti A, Abdollah F. The Impact of Positron Emission Tomography Imaging and Tumor Molecular Profiling on Risk Stratification, Treatment Choice, and Oncological Outcomes of Patients with Primary or Relapsed Prostate Cancer: An International Collaborative Review of the Existing Literature. Eur Urol Oncol 2024; 7:27-43. [PMID: 37423774 DOI: 10.1016/j.euo.2023.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 05/06/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
CONTEXT The clinical introduction of next-generation imaging methods and molecular biomarkers ("radiogenomics") has revolutionized the field of prostate cancer (PCa). While the clinical validity of these tests has thoroughly been vetted, their clinical utility remains a matter of investigation. OBJECTIVE To systematically review the evidence to date on the impact of positron emission tomography (PET) imaging and tissue-based prognostic biomarkers, including Decipher, Prolaris, and Oncotype Dx, on the risk stratification, treatment choice, and oncological outcomes of men with newly diagnosed PCa or those with biochemical failure (BCF). EVIDENCE ACQUISITION We performed a quantitative systematic review of the literature using the MEDLINE, EMBASE, and Web of Science databases (2010-2022) following the Preferred Reporting Items for Systematic Reviews and Meta-analyses statement guidelines. The validated Quality Assessment of Diagnostic Accuracy Studies 2 scoring system was used to assess the risk of bias. EVIDENCE SYNTHESIS A total of 148 studies (130 on PET and 18 on biomarkers) were included. In the primary PCa setting, prostate-specific membrane antigen (PSMA) PET imaging was not useful in improving T staging, moderately useful in improving N staging, but consistently useful in improving M staging in patients with National Comprehensive Cancer Network (NCCN) unfavorable intermediate- to very-high-risk PCa. Its use led to a management change in 20-30% of patients. However, the effect of these treatment changes on survival outcomes was not clear. Similarly, biomarkers in the pretherapy primary PCa setting increased and decreased the risk, respectively, in 7-30% and 32-36% of NCCN low-risk and 31-65% and 4-15% of NCCN favorable intermediate-risk patients being considered for active surveillance. A change in management was noted in up to 65% of patients, with the change being in line with the molecular risk-based reclassification, but again, the impact of these changes on survival outcomes remained unclear. Notably, in the postsurgical primary PCa setting, biomarker-guided adjuvant radiation therapy (RT) was associated with improved oncological control: Δ↓ 2-yr BCF by 22% (level 2b). In the BCF setting, the data were more mature. PSMA PET was consistently useful in improving disease localization-Δ↑ detection for T, N, and M staging was 13-32%, 19-58%, and 9-29%, respectively. Between 29% and 73% of patients had a change in management. Most importantly, these management changes were associated with improved survival outcomes in three trials: Δ↑ 4-yr disease-free survival by 24.3%, Δ↑ 6-mo metastasis-free survival (MFS) by 46.7%, and Δ↑ androgen deprivation therapy-free survival by 8 mo in patients who received PET-concordant RT (level 1b-2b). Biomarker testing in these patients also appeared to be helpful in risk stratifying and guiding the use of early salvage RT (sRT) and concomitant hormonal therapy. Patients with high-genomic-risk scores benefitted from treatment intensification: Δ↑ 8-yr MFS by 20% with the use of early sRT and Δ↑ 12-yr MFS by 11.2% with the use of hormonal therapy alongside early sRT, while low-genomic-risk score patients did equally well with initial conservative management (level 3). CONCLUSIONS Both PSMA PET imaging and tumor molecular profiling provide actionable information in the management of men with primary PCa and those with BCF. Emerging data suggest that radiogenomics-guided treatments translate into direct survival benefits for patients, however, additional prospective data are awaited. PATIENT SUMMARY In this review, we evaluated the utility of prostate-specific membrane antigen positron emission tomography and tumor molecular profiling in guiding the care of men with prostate cancer (PCa). We found that these tests augmented risk stratification, altered management, and improved cancer control in men with a new diagnosis of PCa or for those experiencing a relapse.
Collapse
Affiliation(s)
- Akshay Sood
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Urology, The James Cancer Hospital and Solove Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Amar U Kishan
- Department of Radiation Oncology and Urology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Christopher P Evans
- Department of Urologic Surgery, University of California Davis, Sacramento, CA, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Todd M Morgan
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Declan G Murphy
- Department of Genitourinary Oncology, Peter MacCallum Cancer Centre, The University of Melbourne, Victoria, Australia
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| | - Peter Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henk G Van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 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; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Alberto Briganti
- Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Firas Abdollah
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
16
|
Maekawa S, Takata R, Obara W. Molecular Mechanisms of Prostate Cancer Development in the Precision Medicine Era: A Comprehensive Review. Cancers (Basel) 2024; 16:523. [PMID: 38339274 PMCID: PMC10854717 DOI: 10.3390/cancers16030523] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
The progression of prostate cancer (PCa) relies on the activation of the androgen receptor (AR) by androgens. Despite efforts to block this pathway through androgen deprivation therapy, resistance can occur through several mechanisms, including the abnormal activation of AR, resulting in castration-resistant PCa following the introduction of treatment. Mutations, amplifications, and splicing variants in AR-related genes have garnered attention in this regard. Furthermore, recent large-scale next-generation sequencing analysis has revealed the critical roles of AR and AR-related genes, as well as the DNA repair, PI3K, and cell cycle pathways, in the onset and progression of PCa. Moreover, research on epigenomics and microRNA has increasingly become popular; however, it has not translated into the development of effective therapeutic strategies. Additionally, treatments targeting homologous recombination repair mutations and the PI3K/Akt pathway have been developed and are increasingly accessible, and multiple clinical trials have investigated the efficacy of immune checkpoint inhibitors. In this comprehensive review, we outline the status of PCa research in genomics and briefly explore potential future developments in the field of epigenetic modifications and microRNAs.
Collapse
Affiliation(s)
- Shigekatsu Maekawa
- Department of Urology, Iwate Medical University, Iwate 028-3694, Japan; (R.T.); (W.O.)
| | | | | |
Collapse
|
17
|
Sumiyoshi T, Wang X, Warner EW, Sboner A, Annala M, Sigouros M, Beja K, Mizuno K, Ku S, Fazli L, Eastham J, Taplin ME, Simko J, Halabi S, Morris MJ, Gleave ME, Wyatt AW, Beltran H. Molecular features of prostate cancer after neoadjuvant therapy in the phase 3 CALGB 90203 trial. J Natl Cancer Inst 2024; 116:115-126. [PMID: 37676819 PMCID: PMC10777679 DOI: 10.1093/jnci/djad184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/31/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND The phase 3 CALGB 90203 (Alliance) trial evaluated neoadjuvant chemohormonal therapy for high-risk localized prostate cancer before radical prostatectomy. We dissected the molecular features of post-treated tumors with long-term clinical outcomes to explore mechanisms of response and resistance to chemohormonal therapy. METHODS We evaluated 471 radical prostatectomy tumors, including 294 samples from 166 patients treated with 6 cycles of docetaxel plus androgen deprivation therapy before radical prostatectomy and 177 samples from 97 patients in the control arm (radical prostatectomy alone). Targeted DNA sequencing and RNA expression of tumor foci and adjacent noncancer regions were analyzed in conjunction with pathologic changes and clinical outcomes. RESULTS Tumor fraction estimated from DNA sequencing was significantly lower in post-treated tumor tissues after chemohormonal therapy compared with controls. Higher tumor fraction after chemohormonal therapy was associated with aggressive pathologic features and poor outcomes, including prostate-specific antigen-progression-free survival. SPOP alterations were infrequently detected after chemohormonal therapy, while TP53 alterations were enriched and associated with shorter overall survival. Residual tumor fraction after chemohormonal therapy was linked to higher expression of androgen receptor-regulated genes, cell cycle genes, and neuroendocrine genes, suggesting persistent populations of active prostate cancer cells. Supervised clustering of post-treated high-tumor-fraction tissues identified a group of patients with elevated cell cycle-related gene expression and poor clinical outcomes. CONCLUSIONS Distinct recurrent prostate cancer genomic and transcriptomic features are observed after exposure to docetaxel and androgen deprivation therapy. Tumor fraction assessed by DNA sequencing quantifies pathologic response and could be a useful trial endpoint or prognostic biomarker. TP53 alterations and high cell cycle transcriptomic activity are linked to aggressive residual disease, despite potent chemohormonal therapy.
Collapse
Affiliation(s)
- Takayuki Sumiyoshi
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xiaofei Wang
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Evan W Warner
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Matti Annala
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kevin Beja
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Kei Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shengyu Ku
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ladan Fazli
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - James Eastham
- Urology Service at the Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jeffrey Simko
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke Cancer Institute, Durham, NC, USA
| | - Michael J Morris
- Department of Genitourinary Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin E Gleave
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
18
|
Warner EW, Van der Eecken K, Murtha AJ, Kwan EM, Herberts C, Sipola J, Ng SWS, Chen XE, Fonseca NM, Ritch E, Schönlau E, Bernales CQ, Donnellan G, Munzur AD, Parekh K, Beja K, Wong A, Verbeke S, Lumen N, Van Dorpe J, De Laere B, Annala M, Vandekerkhove G, Ost P, Wyatt AW. Multiregion sampling of de novo metastatic prostate cancer reveals complex polyclonality and augments clinical genotyping. NATURE CANCER 2024; 5:114-130. [PMID: 38177459 DOI: 10.1038/s43018-023-00692-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/15/2023] [Indexed: 01/06/2024]
Abstract
De novo metastatic prostate cancer is highly aggressive, but the paucity of routinely collected tissue has hindered genomic stratification and precision oncology. Here, we leveraged a rare study of surgical intervention in 43 de novo metastatic prostate cancers to assess somatic genotypes across 607 synchronous primary and metastatic tissue regions plus circulating tumor DNA. Intra-prostate heterogeneity was pervasive and impacted clinically relevant genes, resulting in discordant genotypes between select primary restricted regions and synchronous metastases. Additional complexity was driven by polyclonal metastatic seeding from phylogenetically related primary populations. When simulating clinical practice relying on a single tissue region, genomic heterogeneity plus variable tumor fraction across samples caused inaccurate genotyping of dominant disease; however, pooling extracted DNA from multiple biopsy cores before sequencing can rescue misassigned somatic genotypes. Our results define the relationship between synchronous treatment-sensitive primary and metastatic lesions in men with de novo metastatic prostate cancer and provide a framework for implementing genomics-guided patient management.
Collapse
Affiliation(s)
- Evan W Warner
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kim Van der Eecken
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Andrew J Murtha
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Edmond M Kwan
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Cameron Herberts
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joonatan Sipola
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Sarah W S Ng
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xinyi E Chen
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicolette M Fonseca
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elie Ritch
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elena Schönlau
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cecily Q Bernales
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gráinne Donnellan
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aslı D Munzur
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karan Parekh
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Beja
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amanda Wong
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sofie Verbeke
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Nicolaas Lumen
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Bram De Laere
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Matti Annala
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Gillian Vandekerkhove
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Piet Ost
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Alexander W Wyatt
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada.
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada.
| |
Collapse
|
19
|
Wang R, Xu Q, Guo H, Yang G, Zhang J, Wang H, Xu T, Guo C, Yuan J, He Y, Zhang X, Fu H, Xu G, Zhao B, Xie J, Zhao T, Huang L, Zhang J, Peng B, Yao X, Yang B. Concordance and Clinical Significance of Genomic Alterations in Progressive Tumor Tissue and Matched Circulating Tumor DNA in Aggressive-variant Prostate Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:2221-2232. [PMID: 37877742 PMCID: PMC10624154 DOI: 10.1158/2767-9764.crc-23-0175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/09/2023] [Accepted: 10/18/2023] [Indexed: 10/26/2023]
Abstract
Sequencing of circulating tumor DNA (ctDNA) is a minimally invasive approach to reveal the genomic alterations of cancer; however, its comparison with sequencing of tumor tissue has not been well documented in real-world patients with aggressive-variant prostate cancer (AVPC). Concordance of genomic alterations was assessed between progressive tumor tissue and matched ctDNA by next-generation sequencing for 63 patients with AVPC. Associations of genomic alterations with progression-free survival (PFS) and overall survival (OS) were investigated using Kaplan-Meier and Cox regression analyses. A total of 161 somatic mutations (SMs) and 84 copy-number variants (CNVs) were detected in tumors, of which 97 were also found in ctDNA, giving concordance of 39.6% (97/245) across all SMs and CNVs, 49.7% for SMs only and 20.2% for CNVs only. Across all patients with AVPC, chemotherapy was associated with significantly longer median PFS (6 vs. 0.75 months, P = 0.001) and OS (11 vs. 8 months, P < 0.001) than next-generation hormonal therapy (NHT). Among types of chemotherapy, additional platinum-based chemotherapy was associated with significantly longer median PFS and OS than docetaxel only in patients with TP53, RB1, or PTEN alterations, and in those with ctDNA% ≥ 13.5%. The concordance analysis first provides evidence for combining the sequencing of ctDNA and tumor tissue in real-world patients with AVPC. Chemotherapy is associated with significantly better survival than NHT, and the benefit of additional platinum-based chemotherapy may depend on the presence of alterations in TP53, RB1, or PTEN and on a sufficiently high proportion of ctDNA in patients with AVPC. SIGNIFICANCE AVPC is a highly malignant and heterogeneous disease. Sequencing of ctDNA is a minimally invasive approach to reveal genomic alterations. On the basis of the current real-world study, we found ctDNA does not fully recapitulate the landscape of genomic alterations from progressive tumor tissue in AVPC. We also revealed AVPC can benefit from chemotherapy, especially platinum-based regimens. TP53/RB1/PTEN alterations in ctDNA or tumor tissue could be biomarkers for platinum-based chemotherapy in this setting.
Collapse
Affiliation(s)
- Ruiliang Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Qiufan Xu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Hanxu Guo
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Guanjie Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jun Zhang
- Department of Urology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Hong Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Tianyuan Xu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Changcheng Guo
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jing Yuan
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
| | - Yanyan He
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Xiaoying Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Hongliang Fu
- Department of Nuclear Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Guang Xu
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Binghui Zhao
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jun Xie
- Department of Urology, Shanghai Clinical College, Anhui Medical University, Shanghai, P.R. China
| | - Tingting Zhao
- Research Institute, GloriousMed Clinical Laboratory, Shanghai, P.R. China
| | - Longfei Huang
- Research Institute, GloriousMed Clinical Laboratory, Shanghai, P.R. China
| | - Jiansheng Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, P.R. China
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
- Department of Urology, Shanghai Clinical College, Anhui Medical University, Shanghai, P.R. China
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, P.R. China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
- Department of Urology, Shanghai Clinical College, Anhui Medical University, Shanghai, P.R. China
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, P.R. China
| | - Bin Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, P.R. China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, P.R. China
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
20
|
Zaffaroni M, Vincini MG, Corrao G, Lorubbio C, Repetti I, Mastroleo F, Putzu C, Villa R, Netti S, D’Ecclesiis O, Luzzago S, Mistretta FA, Musi G, Cattani F, Gandini S, Marvaso G, Jereczek-Fossa BA. Investigating Nutritional and Inflammatory Status as Predictive Biomarkers in Oligoreccurent Prostate Cancer-A RADIOSA Trial Preliminary Analysis. Nutrients 2023; 15:4583. [PMID: 37960236 PMCID: PMC10647217 DOI: 10.3390/nu15214583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
(1) Background: In the RADIOSA phase II randomized clinical trial (NCT03940235), the biology task entails the identification of predictive and prognostic biomarkers in the context of oligorecurrent, castration-sensitive prostate cancer in order to distinguish polymetastatic from oligometastatic disease. This may lay the groundwork for personalized treatments for those patients who could really benefit from metastasis-directed therapies. (2) Methods: Oligorecurrent PCa pts with three or fewer bone or lymph nodal localizations were randomized 1:1 to receive SBRT alone (arm A) or SBRT + 6 months of ADT (arm B). Common serum-derived biomarkers were collected at baseline, and at 3 months after RT. The prognostic nutritional index, an immune and nutrition-based prognostic score, and the controlling nutritional status (CONUT) score, a scoring system for evaluating patient's nutritional status, were calculated in accordance with the body of available literature. As inflammatory indicators, neutrophil-lymphocyte ratio (NLR) and the NLR-albumin ratio (NLRAR) were assessed. Changes in these parameters between baseline and the 3-month timepoint were evaluated both in absolute and relative values. Changes in these parameters between baseline and the 3-month timepoint were evaluated. Significant differences in the trend of these parameters were assessed using the non-parametric Wilcoxon rank-sum test. A network analysis to analyze the relationships between different features stratifying patients according to the arm of study and site of metastases was performed. (3) Results: The current analysis comprised 88 patients (45 arm A, SBRT only, and 43 arm B, SBRT + ADT). When patients were stratified by ADT administration, cholesterol values showed an increasing trend in the group receiving ADT (p = 0.005) which was no longer significant at 1 year. When patients were stratified by site of metastases (52 lymph nodal, 29 bone localizations), the value of NLR was found to be increased in patients with bone localizations (p < 0.05). In addition, the network analysis showed that BMI and NRI are strongly and directly linked for patients at baseline and that this correlation is no longer found at three months. Finally, when patients were divided according to time from surgery to oligorecurrence (enrollment) the patients with a longer time (>6.7 years) showed an increase in CONUT score from baseline. All the other nutritional and inflammatory scores or parameters investigated in the present analysis showed no statistically significant differences at baseline, three months, 1 year, and in absolute change. (4) Conclusions: The nutritional and inflammatory parameters do not seem to represent valuable candidates for possible use in clinical decision making in our cohort of patients and a reliable biological characterization of the oligometastatic state in prostate cancer still seems far from being achieved. Ongoing molecular analysis will show if there is a role of mutational landscape in the definition of the oligometastatic state.
Collapse
Affiliation(s)
- Mattia Zaffaroni
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
| | - Maria Giulia Vincini
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
| | - Giulia Corrao
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
| | - Chiara Lorubbio
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
| | - Ilaria Repetti
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
| | - Federico Mastroleo
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
- Department of Translational Medicine, University of Piemonte Orientale, 20188 Novara, Italy
| | - Costantino Putzu
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
| | - Riccardo Villa
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
| | - Sofia Netti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (S.N.); (O.D.); (S.G.)
| | - Oriana D’Ecclesiis
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (S.N.); (O.D.); (S.G.)
| | - Stefano Luzzago
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
- Division of Urology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesco Alessandro Mistretta
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
- Division of Urology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Gennaro Musi
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
- Division of Urology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Federica Cattani
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Sara Gandini
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (S.N.); (O.D.); (S.G.)
| | - Giulia Marvaso
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy (M.G.V.); (G.C.); (I.R.); (F.M.); (C.P.); (R.V.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (S.L.); (F.A.M.); (G.M.)
| |
Collapse
|
21
|
Piombino C, Oltrecolli M, Tonni E, Pirola M, Matranga R, Baldessari C, Pipitone S, Dominici M, Sabbatini R, Vitale MG. De Novo Metastatic Prostate Cancer: Are We Moving toward a Personalized Treatment? Cancers (Basel) 2023; 15:4945. [PMID: 37894312 PMCID: PMC10605467 DOI: 10.3390/cancers15204945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
De novo metastatic hormone-sensitive PC (mHSPC) accounts for 5-10% of all prostate cancer (PC) diagnoses but it is responsible for nearly 50% of PC-related deaths. Since 2015, the prognosis of mHSPC has slightly improved thanks to the introduction of new hormonal agents and chemotherapy combined with androgen deprivation therapy from the first-line setting. This review describes the current therapeutic opportunities for de novo mHSPC, focusing on potential molecular biomarkers identified in the main clinical trials that have modified the standard of care, the genomic features of de novo mHSPC, and the principal ongoing trials that are investigating new therapeutic approaches and the efficacy of a biomarker-guided treatment in this setting. The road toward personalized treatment for de novo mHSPC is still long, considering that the randomized clinical trials, which have furnished the basis of the current therapeutic options, stratified patients according to clinical criteria that did not necessarily reflect the biological rationale of the chosen therapy. The role of transcriptomic profiling of mHSPC as a predictive biomarker requires further validation, and it remains to be ascertained how the genomic variants detected in mHSPC, which are regarded as predictive in the castration-resistant disease, can be exploited in the mHSPC setting.
Collapse
Affiliation(s)
- Claudia Piombino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Marco Oltrecolli
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Elena Tonni
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Marta Pirola
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Rossana Matranga
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Cinza Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Stefania Pipitone
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Roberto Sabbatini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| | - Maria Giuseppa Vitale
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (M.O.); (E.T.); (M.P.); (R.M.); (C.B.); (S.P.); (M.D.); (R.S.)
| |
Collapse
|
22
|
Kafka M, Burtscher T, Fritz J, Schmitz M, Bektic J, Ladurner M, Horninger W, Heidegger I. Real-world comparison of Docetaxel versus new hormonal agents in combination with androgen-deprivation therapy in metastatic hormone-sensitive prostate cancer descrying PSA Nadir ≤ 0.05 ng/ml as marker for treatment response. World J Urol 2023; 41:2043-2050. [PMID: 36287244 PMCID: PMC10415491 DOI: 10.1007/s00345-022-04189-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022] Open
Abstract
PROPOSE Using Docetaxel chemotherapy or new hormonal agents (NHT) to intensify upfront systemic therapy resulted in improved survival rates compared to androgen deprivation monotherapy (ADT). Hence, combination therapies have become the new standard of care (SOC) in metastatic hormone-sensitive prostate cancer (mHSPC). However, head-to-head trails comparing different therapies as well as treatment-guiding biomarkers are still lacking. Thus, the aim of the present study was to compare clinical outcomes of Docetaxel versus NHT therapy in the real-world setting as well as to elaborate biomarkers predicting clinical outcome. METHODS We retrospectively assessed overall-survival (OS), progression-free survival 1 and 2 (PFS1/2) and time to progression (TTP) in 42 patients treated by either ADT + NHT or ADT + Docetaxel. In addition, we investigated clinical prognostic biomarkers. RESULTS Our survival analysis revealed 3-year OS of 89.4% in the NHT group compared to 82.4% in the Docetaxel group. 3-year PFS1 was 59.6% in the NHT group compared to 32.2% in the Docetaxel group and the TTP was 53.8% vs 32.2% (pOS = 0.189; pPFS1 = 0.082; pTTP = 0.055). In addition, castration-resistance occurred more often in the Docetaxel group (78.6% vs 25%, p = 0.004). Interestingly, a PSA-Nadir ≤ 0.05 ng/ml during therapy was associated with increased survival rates (p < 0.001) while PSA levels at primary diagnosis had no influence on therapy outcome. Furthermore, a thyroid-stimulating hormone (TSH) increase during therapy was associated with improved clinical outcome (p = 0.06). CONCLUSION We observed a trend towards a higher benefit of NHT as first-line treatment compared to Docetaxel in men with mHSPC. Of note, a PSA-Nadir ≤ 0.05 ng/ml or a TSH-increase during therapy were predictors for therapy response.
Collapse
Affiliation(s)
- Mona Kafka
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas Burtscher
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - Josef Fritz
- Department of Medical Statistics, Informatics and Health Economics, Medical University Innsbruck, Innsbruck, Austria
| | | | - Jasmin Bektic
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - Michael Ladurner
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Isabel Heidegger
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
23
|
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: 1.5] [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
|
24
|
Special Issue "Prostate Cancer: Recent Advances in Diagnostics and Treatment Planning". J Clin Med 2022; 11:jcm11226823. [PMID: 36431300 PMCID: PMC9697296 DOI: 10.3390/jcm11226823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
This editorial of the Special Issue "Prostate Cancer: Recent Advances in Diagnostics and Treatment Planning" aims to draw more attention to the broad and diverse field of prostate cancer (PCa) diagnosis and the utilization of different diagnostic means to improve clinical decision-making and treatment strategy planning [...].
Collapse
|
25
|
De Laere B, Crippa A, Discacciati A, Larsson B, Oldenburg J, Mortezavi A, Ost P, Eklund M, Lindberg J, Grönberg H. Clinical Trial Protocol for ProBio: An Outcome-adaptive and Randomised Multiarm Biomarker-driven Study in Patients with Metastatic Prostate Cancer. Eur Urol Focus 2022; 8:1617-1621. [PMID: 35317973 DOI: 10.1016/j.euf.2022.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/08/2022] [Indexed: 01/25/2023]
Abstract
ProBio is an outcome-adaptive, multiarm, multiple-assignment randomised, biomarker-driven platform trial in men with metastatic castration-resistant prostate cancer. Here we describe the amended clinical protocol, focusing on expansion of the trial to include patients with de novo metastatic hormone-sensitive prostate cancer.
Collapse
Affiliation(s)
- Bram De Laere
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Alessio Crippa
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Discacciati
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Berit Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jan Oldenburg
- Department of Oncology, Akershus University Hospital, Ahus, Norway
| | - Ashkan Mortezavi
- Department of Urology, University Hospital Basel, Basel, Switzerland
| | - Piet Ost
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Department of Radiation Oncology, GZA Sint-Augustinus, Antwerp, Belgium
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Oncology, Capio S:t Görans Sjukhus, Stockholm, Sweden
| |
Collapse
|
26
|
Deek MP, Van der Eecken K, Sutera P, Deek RA, Fonteyne V, Mendes AA, Decaestecker K, Kiess AP, Lumen N, Phillips R, De Bruycker A, Mishra M, Rana Z, Molitoris J, Lambert B, Delrue L, Wang H, Lowe K, Verbeke S, Van Dorpe J, Bultijnck R, Villeirs G, De Man K, Ameye F, Song DY, DeWeese T, Paller CJ, Feng FY, Wyatt A, Pienta KJ, Diehn M, Bentzen SM, Joniau S, Vanhaverbeke F, De Meerleer G, Antonarakis ES, Lotan TL, Berlin A, Siva S, Ost P, Tran PT. Long-Term Outcomes and Genetic Predictors of Response to Metastasis-Directed Therapy Versus Observation in Oligometastatic Prostate Cancer: Analysis of STOMP and ORIOLE Trials. J Clin Oncol 2022; 40:3377-3382. [PMID: 36001857 PMCID: PMC10166371 DOI: 10.1200/jco.22.00644] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/02/2022] [Accepted: 07/21/2022] [Indexed: 11/20/2022] Open
Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported.The initial STOMP and ORIOLE trial reports suggested that metastasis-directed therapy (MDT) in oligometastatic castration-sensitive prostate cancer (omCSPC) was associated with improved treatment outcomes. Here, we present long-term outcomes of MDT in omCSPC by pooling STOMP and ORIOLE and assess the ability of a high-risk mutational signature to risk stratify outcomes after MDT. The primary end point was progression-free survival (PFS) calculated using the Kaplan-Meier method. High-risk mutations were defined as pathogenic somatic mutations within ATM, BRCA1/2, Rb1, or TP53. The median follow-up for the whole group was 52.5 months. Median PFS was prolonged with MDT compared with observation (pooled hazard ratio [HR], 0.44; 95% CI, 0.29 to 0.66; P value < .001), with the largest benefit of MDT in patients with a high-risk mutation (HR high-risk, 0.05; HR no high-risk, 0.42; P value for interaction: .12). Within the MDT cohort, the PFS was 13.4 months in those without a high-risk mutation, compared with 7.5 months in those with a high-risk mutation (HR, 0.53; 95% CI, 0.25 to 1.11; P = .09). Long-term outcomes from the only two randomized trials in omCSPC suggest a sustained clinical benefit to MDT over observation. A high-risk mutational signature may help risk stratify treatment outcomes after MDT.
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
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kim Van der Eecken
- Department of Pathology and Human Structure and Repair, University of Ghent, Ghent, Belgium
| | - Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rebecca A. Deek
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Valérie Fonteyne
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Adrianna A. Mendes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Ana Ponce Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicolaas Lumen
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Ryan Phillips
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | | | - Mark Mishra
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | - Zaker Rana
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | - Jason Molitoris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | - Bieke Lambert
- Department of Radiology and Nuclear Medicine, Ghent University, and Department of Nuclear Medicine, AZ Maria-Middelares Ghent, Belgium
| | - Louke Delrue
- Department of Radiology, Ghent University Hospital, Ghent, Belgium
| | - Hailun Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kathryn Lowe
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sofie Verbeke
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Renée Bultijnck
- Department of Urology, Ghent University Hospital, Ghent, Belgium
| | - Geert Villeirs
- Department of Radiology and Nuclear Medicine, Ghent University, and Department of Nuclear Medicine, AZ Maria-Middelares Ghent, Belgium
| | - Kathia De Man
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Filip Ameye
- Department of Urology, AZ Maria-Middelares Ghent, Ghent, Belgium
| | - Daniel Y. Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Theodore DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Channing J. Paller
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Felix Y. Feng
- Departments of Medicine, Urology and Radiation Oncology, UCSF, San Francisco, CA
| | - Alexander Wyatt
- Department of Urologic Sciences, University of British Columbia, and Vancouver Prostate Centre, Vancouver, Canada
| | - Kenneth J. Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD
| | - Maximillian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Soren M. Bentzen
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD
| | - Steven Joniau
- Department of Urology, Catholic University Leuven, Leuven, Belgium
| | | | - Gert De Meerleer
- Department of Radiation Oncology, Catholic University Leuven, Leuven, Belgium
| | | | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alejandro Berlin
- Department of Radiation Oncology, Princess Margaret Cancer Center, Toronto, Canada
| | - Shankar Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Center, Melbourne Australia
| | - 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, MD
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
27
|
Genomic Landscape Alterations in Primary Tumor and Matched Lymph Node Metastasis in Hormone-Naïve Prostate Cancer Patients. Cancers (Basel) 2022; 14:cancers14174212. [PMID: 36077746 PMCID: PMC9454441 DOI: 10.3390/cancers14174212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Prostate cancer (PCa) is a disease with a wide range of clinical manifestations. Up to the present date, the genetic understanding of patients with favorable or unfavorable prognosis is gaining interest for giving the appropriate tailored treatment. We aimed to investigate genetic changes associated with lymph node metastasis in a cohort of hormone-naïve Pca patients. Methods: We retrospectively analyzed data from 470 patients who underwent surgery for PCa between 2010 and 2020 at the Department of Urology, University of Catania. Inclusion criteria were patients with lymph node metastasis and patients with PCa with extra capsular extension (pT3) and negative lymph node metastasis. The final cohort consisted of 17 different patients (11 PCa with lymph node metastasis and 6 PCa without lymph node metastasis). Through the cBioPortal online tool, we analyzed gene alterations and their correlations with clinical factors. Results: A total of 688 intronic, synonym and nonsynonym mutations were sequenced. The gene with the most sequenced mutations was ERBB4 (83 mutations, 12% of 688 total), while the ones with the lower percentage of mutations were AKT1, FGFR2 and MLH1 (1 mutation alone, 0.14%). Conclusion: In the present study we found mostly concordance concerning the ERBB4 mutation between both primary PCa samples and matched lymph node metastasis, underlining that the identification of alterations in the primary tumor is extremely important for cancer prognosis prediction.
Collapse
|
28
|
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: 3] [Impact Index Per Article: 1.0] [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
|
29
|
Choudhury AD. PTEN-PI3K pathway alterations in advanced prostate cancer and clinical implications. Prostate 2022; 82 Suppl 1:S60-S72. [PMID: 35657152 DOI: 10.1002/pros.24372] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/21/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite significant advances in molecular characterization and therapeutic targeting of advanced prostate cancer, it remains the second most common cause of cancer death in men in the United States. The PI3K (Phosphatidylinositol 3-kinase)/AKT (AKT serine/threonine kinase)/mTOR (mammalian target of rapamycin) signaling pathway is commonly altered in prostate cancer, most frequently through loss of the PTEN (Phosphatase and Tensin Homolog) tumor suppressor, and is critical for cancer cell proliferation, migration, and survival. METHODS This study summarizes signaling through the PTEN/PI3K pathway, alterations in pathway components commonly seen in advanced prostate cancer, and results of clinical trials of pathway inhibitors reported to date with a focus on more recently reported studies. It also reviews rationale for combination approaches currently under study, including with taxanes, immune checkpoint inhibitors and poly (ADP-ribose) polymerase inhibitors, and discusses future directions in biomarker testing and therapeutic targeting of this pathway. RESULTS Clinical trials studying pharmacologic inhibitors of PI3K, AKT or mTOR kinases have demonstrated modest activity of specific agents, with several trials of pathway inhibitors currently in progress. A key challenge is the importance of PI3K/AKT/mTOR signaling in noncancerous tissues, leading to predictable but often severe toxicities at therapeutic doses. RESULTS Further advances in selective pharmacologic inhibition of the PI3K/AKT/mTOR pathway in tumors, development of rational combinations, and appropriate biomarker selection to identify the appropriate tumor- and patient-specific vulnerabilities will be required to optimize clinical benefit from therapeutic targeting of this pathway.
Collapse
Affiliation(s)
- Atish D Choudhury
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Abstract
In recent years, wearable sensors have enabled the unique mode of real-time and noninvasive monitoring to develop rapidly in medical care, sports, and other fields. Sweat contains a wide range of biomarkers such as metabolites, electrolytes, and various hormones. Combined with wearable technology, sweat can reflect human fatigue, disease, mental stress, dehydration, and so on. This paper comprehensively describes the analysis of sweat components such as glucose, lactic acid, electrolytes, pH, cortisol, vitamins, ethanol, and drugs by wearable sensing technology, and the application of sweat wearable devices in glasses, patches, fabrics, tattoos, and paper. The development trend of sweat wearable devices is prospected. It is believed that if the sweat collection, air permeability, biocompatibility, sensing array construction, continuous monitoring, self-healing technology, power consumption, real-time data transmission, specific recognition, and other problems of the wearable sweat sensor are solved, we can provide the wearer with important information about their health level in the true sense.
Collapse
|
31
|
Nevedomskaya E, Haendler B. From Omics to Multi-Omics Approaches for In-Depth Analysis of the Molecular Mechanisms of Prostate Cancer. Int J Mol Sci 2022; 23:6281. [PMID: 35682963 PMCID: PMC9181488 DOI: 10.3390/ijms23116281] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/24/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer arises following alterations at different cellular levels, including genetic and epigenetic modifications, transcription and translation dysregulation, as well as metabolic variations. High-throughput omics technologies that allow one to identify and quantify processes involved in these changes are now available and have been instrumental in generating a wealth of steadily increasing data from patient tumors, liquid biopsies, and from tumor models. Extensive investigation and integration of these data have led to new biological insights into the origin and development of multiple cancer types and helped to unravel the molecular networks underlying this complex pathology. The comprehensive and quantitative analysis of a molecule class in a biological sample is named omics and large-scale omics studies addressing different prostate cancer stages have been performed in recent years. Prostate tumors represent the second leading cancer type and a prevalent cause of cancer death in men worldwide. It is a very heterogenous disease so that evaluating inter- and intra-tumor differences will be essential for a precise insight into disease development and plasticity, but also for the development of personalized therapies. There is ample evidence for the key role of the androgen receptor, a steroid hormone-activated transcription factor, in driving early and late stages of the disease, and this led to the development and approval of drugs addressing diverse targets along this pathway. Early genomic and transcriptomic studies have allowed one to determine the genes involved in prostate cancer and regulated by androgen signaling or other tumor-relevant signaling pathways. More recently, they have been supplemented by epigenomic, cistromic, proteomic and metabolomic analyses, thus, increasing our knowledge on the intricate mechanisms involved, the various levels of regulation and their interplay. The comprehensive investigation of these omics approaches and their integration into multi-omics analyses have led to a much deeper understanding of the molecular pathways involved in prostate cancer progression, and in response and resistance to therapies. This brings the hope that novel vulnerabilities will be identified, that existing therapies will be more beneficial by targeting the patient population likely to respond best, and that bespoke treatments with increased efficacy will be available soon.
Collapse
Affiliation(s)
| | - Bernard Haendler
- Research and Early Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany;
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW To summarize the role of chemotherapy and offer some guidance regarding the selection of chemotherapy in mPC. RECENT FINDINGS Patients with mHSPC have varied prognoses with testosterone suppression alone (androgen deprivation therapy, ADT) and differential responses to docetaxel with ADT. Patients with de novo and metachronous high-volume disease have a robust survival benefit with the addition of docetaxel to hormonal therapies. Patients with synchronous low-volume disease have a more modest survival benefit from docetaxel and there is no evidence of survival benefit with docetaxel in patients with metachronous low-volume disease. Integration of biomarkers may refine treatment selection regardless of volume of disease. Docetaxel and cabazitaxel also impart an OS benefit in patients with metastatic castration-resistant prostate cancer (mCRPC). The choice of chemotherapy in mCRPC depends on treatment received in mHSPC setting. Docetaxel remains the first line chemotherapy in castration-resistant patients who have not received it in mHSPC followed by cabazitaxel, otherwise cabazitaxel can be deployed without docetaxel retreatment. SUMMARY Chemotherapy is a key class of therapy for selected patients with mHSPC and mCRPC.
Collapse
Affiliation(s)
- Irbaz B Riaz
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Brigham and Women Hospital, Harvard Medical, Massachusetts
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic, Arizona, USA
| | - Christopher J Sweeney
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School
- Brigham and Women Hospital, Harvard Medical, Massachusetts
| |
Collapse
|
33
|
Papachristodoulou A, Abate-Shen C. Precision intervention for prostate cancer: Re-evaluating who is at risk. Cancer Lett 2022; 538:215709. [DOI: 10.1016/j.canlet.2022.215709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/30/2022] [Accepted: 04/25/2022] [Indexed: 02/08/2023]
|
34
|
Beltran H, Choudhury AD. Towards Biologically Driven Decision-making in Metastatic Hormone-sensitive Prostate Cancer. Eur Urol Oncol 2021; 4:924-926. [PMID: 34857503 DOI: 10.1016/j.euo.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
| | - Atish D Choudhury
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|