1
|
Lenz L, Clegg W, Iliev D, Kasten CR, Korman H, Morgan TM, Hafron J, DeHaan A, Olsson C, Tutrone RF, Richardson T, Cline K, Yonover PM, Jasper J, Cohen T, Finch R, Slavin TP, Gutin A. Active surveillance selection and 3-year durability in intermediate-risk prostate cancer following genomic testing. Prostate Cancer Prostatic Dis 2025; 28:427-434. [PMID: 39237680 DOI: 10.1038/s41391-024-00888-y] [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: 12/20/2023] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Genomic testing can add risk stratification information to clinicopathological features in prostate cancer, aiding in shared medical decision-making between the clinician and patient regarding whether active surveillance (AS) or definitive treatment (DT) is most appropriate. Here we examined initial AS selection and 3-year AS durability in patients diagnosed with localized intermediate-risk prostate cancer who underwent Prolaris testing before treatment decision-making. METHODS This retrospective observational cohort study included 3208 patients from 10 study sites who underwent Prolaris testing at diagnosis from September 2015 to December 2018. Prolaris utilizes a combined clinical cell cycle risk score calculated at diagnostic biopsy to stratify patients by the Prolaris AS threshold (below threshold, patient recommended to AS or above threshold, patient recommended to DT). AS selection rates and 3-year AS durability were compared in patients recommended to AS or DT by Prolaris testing. Univariable and multivariable logistic regression models and Cox proportional hazard models were used with molecular and clinical variables as predictors of initial treatment decision and AS durability, respectively. RESULTS AS selection was ~2 times higher in patients recommended to AS by Prolaris testing than in those recommended to DT (p < 0.0001). Three-year AS durability was ~1.5 times higher in patients recommended to AS by Prolaris testing than in those recommended to DT (p < 0.0001). Prolaris treatment recommendation remained a statistically significant predictor of initial AS selection and AS durability after accounting for CAPRA or Gleason scores. CONCLUSIONS Prolaris added significant information to clinical risk stratification to aid in treatment decision making. Intermediate-risk prostate cancer patients who were recommended to AS by Prolaris were more likely to initially pursue AS and were more likely to remain on AS at 3 years post-diagnosis than patients recommended to DT.
Collapse
Affiliation(s)
- Lauren Lenz
- Myriad Genetics, Inc., Salt Lake City, UT, USA
| | - Wyatt Clegg
- Myriad Genetics, Inc., Salt Lake City, UT, USA
| | - Diana Iliev
- Myriad Genetics, Inc., Salt Lake City, UT, USA
| | | | - Howard Korman
- Comprehensive Urology, Royal Oak, MI, USA
- Wayne State University, Detroit, MI, USA
| | | | | | | | - Carl Olsson
- Integrated Medical Professionals, Melville, NY, USA
| | | | | | | | | | - Jeff Jasper
- Myriad Genetics, Inc., Salt Lake City, UT, USA
| | - Todd Cohen
- Myriad Genetics, Inc., Salt Lake City, UT, USA
| | | | | | | |
Collapse
|
2
|
Sharma D, Singh V, Kumar A, Singh TG. Genistein: A promising ally in combating neurodegenerative disorders. Eur J Pharmacol 2025; 991:177273. [PMID: 39828018 DOI: 10.1016/j.ejphar.2025.177273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Neurodegenerative disorders arise when nerve cells in the brain or peripheral nervous system gradually lose functions and eventually die. Although certain therapies may alleviate some of the physical and mental symptoms associated with neurodegenerative disorders, hence slowing their progression, but no sure-shot treatment is currently available. It was shown that the rise in life expectancy and the number of elderly people in the community led to an increasing trend in the incidence and prevalence of neurodegenerative disease. Phytomolecules are demonstrating their effectiveness in combating, regression, and delaying various diseases. Genistein is one of soy isoflavone with antioxidant, anti-inflammatory, and estrogenic effects. Researchers demonstrated that Genistein treatment significantly reduced hyperglycemia, improved cognitive performance by modulating acetylcholinesterase activity and oxidative stress, and alleviated neuroinflammatory conditions in mice. This paper evaluates (in vivo and in vitro) various molecular targets of isoflavones and their ability to effectively counter several neurodegenerative disorders such as Parkinson's, Alzheimer's, and Huntington's diseases and amyotrophic lateral sclerosis. In this review, we aim to provide an overview of the role that genistein plays in delaying the development of neurodegenerative disorders.
Collapse
Affiliation(s)
- Diksha Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Varinder Singh
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India.
| | - Amit Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | | |
Collapse
|
3
|
Tward J, Lloyd S, Johnson S, Dechet C, Nei BO, Maughan B, Swami U, Gupta S, Sanchez A, Kokeny K, Agarwal N. A Phase 2 Trial of Radium 223 and Stereotactic Ablative Radiation Therapy in Hormone-Naïve Men with Oligometastatic Prostate Cancer to Bone: The RadSABR Study. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00095-1. [PMID: 39922320 DOI: 10.1016/j.ijrobp.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE We hypothesized that treatment with Radium223 (Ra223) and Stereotactic ablative radiotherapy (SABR) in patients with bone-only metachronous oligometastastic hormone-sensitive prostate cancer (moHSPC) could safely delay the start of androgen deprivation therapy (ADT) and maintain quality of life (QoL). METHODS AND MATERIALS This prospective trial included 20 men with moHSPC and ≤5 bone-only metastases who previously had definitive treatment to the prostate and pelvic lymph nodes. Eligibility criteria were testosterone ≥ 100 ng/dL and metastases validated by conventional imaging. Exclusion criteria were postinitial treatment (LHRH) therapy or N1 disease at bone metastasis diagnosis. Treatment included 6 cycles of Ra223 and SABR (30 Gy in 5 fractions). Bone scans and Prostate Specific Antigen (PSA) levels were monitored regularly. The primary endpoint was freedom from ADT use at 15 months in ≥20% of patients. Patients were followed for 2 years, with clinically significant patient-reported outcome changes defined as >1/2 standard deviation from baseline. Statistical analyses used Wilcoxon rank sum, Pearson's χ2 tests, and univariate Cox regression, with significance set at P < .05 RESULTS: The median number of Ra223 cycles was 6. Freedom from ADT at 15 and 24 months was 50.0% and 40.0%, respectively (P < .001). Eleven (55%) and 5 (25%) patients had PSA declines exceeding 50% and 90%, respectively, with 2 patients achieving undetectable PSA levels (<0.01) at 2 years. No significant changes were observed in any patient-reported outcome QoL domains. Two patients had grade 3 skeletal-related events, and grade 2+ events attributed to Ra223 and SABR were observed in 4 and 2 patients, respectively. CONCLUSIONS In this phase 2 trial, the initial use of Ra223 and SABR for moHSPC significantly delayed ADT use compared with historical controls. The therapy is well tolerated, preserves QoL, and can lead to undetectable PSA levels at 2 years.
Collapse
Affiliation(s)
- Jonathan Tward
- Department of Radiation Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah.
| | - Shane Lloyd
- Department of Radiation Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Skyler Johnson
- Department of Radiation Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Christopher Dechet
- Department of Urologic Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Brock O Nei
- Department of Urologic Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Benjamin Maughan
- Hematology and Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Umang Swami
- Hematology and Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Sumati Gupta
- Hematology and Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Alejandro Sanchez
- Department of Urologic Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Kristine Kokeny
- Department of Radiation Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| | - Neeraj Agarwal
- Hematology and Oncology, Huntsman Cancer Institute at the University of Utah, Salt Lake City, Utah
| |
Collapse
|
4
|
Tward JD, Huang HC, Esteva A, Mohamad O, van der Wal D, Simko JP, DeVries S, Zhang J, Joun S, Showalter TN, Schaeffer EM, Morgan TM, Monson JM, Wallace JA, Bahary JP, Sandler HM, Spratt DE, Rodgers JP, Feng FY, Tran PT. Prostate Cancer Risk Stratification in NRG Oncology Phase III Randomized Trials Using Multimodal Deep Learning With Digital Histopathology. JCO Precis Oncol 2024; 8:e2400145. [PMID: 39447096 PMCID: PMC11520341 DOI: 10.1200/po.24.00145] [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: 03/06/2024] [Revised: 07/08/2024] [Accepted: 08/22/2024] [Indexed: 10/26/2024] Open
Abstract
PURPOSE Current clinical risk stratification methods for localized prostate cancer are suboptimal, leading to over- and undertreatment. Recently, machine learning approaches using digital histopathology have shown superior prognostic ability in phase III trials. This study aims to develop a clinically usable risk grouping system using multimodal artificial intelligence (MMAI) models that outperform current National Comprehensive Cancer Network (NCCN) risk groups. MATERIALS AND METHODS The cohort comprised 9,787 patients with localized prostate cancer from eight NRG Oncology randomized phase III trials, treated with radiation therapy, androgen deprivation therapy, and/or chemotherapy. Locked MMAI models, which used digital histopathology images and clinical data, were applied to each patient. Expert consensus on cut points defined low-, intermediate-, and high-risk groups on the basis of 10-year distant metastasis rates of 3% and 10%, respectively. The MMAI's reclassification and prognostic performance were compared with the three-tier NCCN risk groups. RESULTS The median follow-up for censored patients was 7.9 years. According to NCCN risk categories, 30.4% of patients were low-risk, 25.5% intermediate-risk, and 44.1% high-risk. The MMAI risk classification identified 43.5% of patients as low-risk, 34.6% as intermediate-risk, and 21.8% as high-risk. MMAI reclassified 1,039 (42.0%) patients initially categorized by NCCN. Despite the MMAI low-risk group being larger than the NCCN low-risk group, the 10-year metastasis risks were comparable: 1.7% (95% CI, 0.2 to 3.2) for NCCN and 3.2% (95% CI, 1.7 to 4.7) for MMAI. The overall 10-year metastasis risk for NCCN high-risk patients was 16.6%, with MMAI further stratifying this group into low-, intermediate-, and high-risk, showing metastasis rates of 3.4%, 8.2%, and 26.3%, respectively. CONCLUSION The MMAI risk grouping system expands the population of men identified as having low metastatic risk and accurately pinpoints a high-risk subset with elevated metastasis rates. This approach aims to prevent both overtreatment and undertreatment in localized prostate cancer, facilitating shared decision making.
Collapse
Affiliation(s)
| | | | | | - Osama Mohamad
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA
| | | | - Jeffry P. Simko
- Department of Urology, University of California San Francisco, San Francisco, CA
| | | | | | | | - Timothy N. Showalter
- Artera, Inc, Los Altos, CA
- Department of Radiation Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | | | - Todd M. Morgan
- Division of Urologic Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Jedidiah M. Monson
- Department of Radiation Oncology, Saint Agnes Medical Center, Fresno, CA
| | | | - Jean-Paul Bahary
- Department of Radiation Oncology, CHUM—Centre Hospitalier de l’Universite de Montreal, Montreal, QC, Canada
| | - Howard M. Sandler
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Daniel E. Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Joseph P. Rodgers
- NRG Oncology Statistics and Data Management Center, American College of Radiology, Philadelphia, PA
| | - Felix Y. Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA
| | - Phuoc T. Tran
- Department of Radiation Oncology, University of Maryland, Baltimore, MD
| |
Collapse
|
5
|
Charlton PV, O'Reilly D, Philippou Y, Rao SR, Lamb ADG, Mills IG, Higgins GS, Hamdy FC, Verrill C, Buffa FM, Bryant RJ. Molecular analysis of archival diagnostic prostate cancer biopsies identifies genomic similarities in cases with progression post-radiotherapy, and those with de novo metastatic disease. Prostate 2024; 84:977-990. [PMID: 38654435 PMCID: PMC11253896 DOI: 10.1002/pros.24715] [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: 01/11/2024] [Accepted: 03/18/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND It is important to identify molecular features that improve prostate cancer (PCa) risk stratification before radical treatment with curative intent. Molecular analysis of historical diagnostic formalin-fixed paraffin-embedded (FFPE) prostate biopsies from cohorts with post-radiotherapy (RT) long-term clinical follow-up has been limited. Utilizing parallel sequencing modalities, we performed a proof-of-principle sequencing analysis of historical diagnostic FFPE prostate biopsies. We compared patients with (i) stable PCa (sPCa) postprimary or salvage RT, (ii) progressing PCa (pPCa) post-RT, and (iii) de novo metastatic PCa (mPCa). METHODS A cohort of 19 patients with diagnostic prostate biopsies (n = 6 sPCa, n = 5 pPCa, n = 8 mPCa) and mean 4 years 10 months follow-up (diagnosed 2009-2016) underwent nucleic acid extraction from demarcated malignancy. Samples underwent 3'RNA sequencing (3'RNAseq) (n = 19), nanoString analysis (n = 12), and Illumina 850k methylation (n = 8) sequencing. Bioinformatic analysis was performed to coherently identify differentially expressed genes and methylated genomic regions (MGRs). RESULTS Eighteen of 19 samples provided useable 3'RNAseq data. Principal component analysis (PCA) demonstrated similar expression profiles between pPCa and mPCa cases, versus sPCa. Coherently differentially methylated probes between these groups identified ~600 differentially MGRs. The top 50 genes with increased expression in pPCa patients were associated with reduced progression-free survival post-RT (p < 0.0001) in an external cohort. CONCLUSIONS 3'RNAseq, nanoString and 850k-methylation analyses are each achievable from historical FFPE diagnostic pretreatment prostate biopsies, unlocking the potential to utilize large cohorts of historic clinical samples. Profiling similarities between individuals with pPCa and mPCa suggests biological similarities and historical radiological staging limitations, which warrant further investigation.
Collapse
Affiliation(s)
- Philip Vincent Charlton
- Department of OncologyUniversity of OxfordOxfordUK
- Department of OncologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | | | - Yiannis Philippou
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Srinivasa Rao Rao
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | - Alastair David Gordon Lamb
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | | | - Geoff Stuart Higgins
- Department of OncologyUniversity of OxfordOxfordUK
- Department of OncologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Freddie Charles Hamdy
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | - Clare Verrill
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
- Department of PathologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | | | - Richard John Bryant
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| |
Collapse
|
6
|
Tward JD, Lenz L, Gutin A, Clegg W, Kasten CR, Finch R, Cohen T, Michalski J, Kishan AU. Using the Cell-Cycle Risk Score to Predict the Benefit of Androgen-Deprivation Therapy Added to Radiation Therapy in Patients With Newly Diagnosed Prostate Cancer. JCO Precis Oncol 2024; 8:e2300722. [PMID: 38748970 PMCID: PMC11371120 DOI: 10.1200/po.23.00722] [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: 12/26/2023] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 09/05/2024] Open
Abstract
PURPOSE Guidelines recommend adding androgen-deprivation therapy (ADT) to radiation therapy (RT) in certain patients with localized prostate cancer. Individualized genomic testing may improve the prognostic accuracy of risk assessments. Herein, we describe a mathematical model of the benefit of adding ADT to RT as a function of the personalized clinical cell-cycle risk (CCR) score to inform 10-year metastasis risk. METHODS A model of absolute risk reduction (ARR) was built using a retrospective cohort of men tested with Prolaris who received RT alone (N = 467). The relative benefit of ADT added to RT to reduce distant metastasis was estimated at 41% on the basis of a meta-analysis of randomized trials. The ARR and number needed to treat (NNT) were computationally derived in patients clinically tested with Prolaris between January 1, 2020, and October 31, 2022 (N = 56,485). Risks were predicted using a cause-specific Cox proportional hazards model with CCR score predicting time to metastasis. A CCR score of 2.112 represents the validated multimodal treatment (MMT) threshold. RESULTS The ARR from ADT increased from almost zero at low CCR scores to 17.1% at CCR = 3.690 with the corresponding NNT = 6, indicating that adding ADT to RT would prevent metastasis within 10 years for one of every six treated individuals. In the clinical cohort, the average ARR was 0.86% in individuals under the MMT threshold (NNT = 116). The average ARR was 8.19% in individuals above the MMT threshold (NNT = 12). Broad ranges of ADT benefit were observed within National Comprehensive Cancer Network risk categories. CONCLUSION The precise and personalized risk estimate of metastasis provided by the CCR score can help inform patients and physicians when considering treatment intensification.
Collapse
|
7
|
Hutten RJ, Odei B, Johnson SB, Tward JD. Validation of the Combined Clinical Cell-Cycle Risk Score to Prognosticate Early Prostate Cancer Metastasis From Biopsy Specimens and Comparison With Other Routinely Used Risk Classifiers. JCO Precis Oncol 2024; 8:e2300364. [PMID: 38330260 DOI: 10.1200/po.23.00364] [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: 07/11/2023] [Revised: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 02/10/2024] Open
Abstract
PURPOSE We aim to independently validate the prognostic utility of the combined cell-cycle risk (CCR) multimodality threshold to estimate risk of early metastasis after definitive treatment of prostate cancer and compare this prognostic ability with other validated biomarkers. METHODS Patients diagnosed with localized prostate cancer were enrolled into a single-institutional registry for the prospective observational cohort study. The primary end point was risk of metastasis within 3 years of diagnostic biopsy. Secondary end points included time to definitive treatment, time to subsequent therapy, and metastasis after completion of initial definitive treatment. Multivariable cause-specific Cox proportional hazards regression models were produced accounting for competing risk of death and stratified on the basis of the CCR active surveillance and multimodality (MM) thresholds. Time-dependent areas under the receiver operating characteristic curve were calculated. RESULTS The cohort consisted of 554 men with prostate cancer and available CCR score from biopsy. The CCR score was prognostic for metastasis (hazard ratio [HR], 2.32 [95% CI, 1.17 to 4.59]; P = .02), with scores above the MM threshold having a higher risk than those below the threshold (HR, 5.44 [95% CI, 2.72 to 10.91]; P < .001). The AUC for 3-year risk of metastasis on the basis of CCR was 0.736. When men with CCR above the MM threshold received MM therapy, their 3-year risk of metastasis was significantly lower than those receiving single-modality therapy (3% v 14%). Similarly, a CCR score above the active surveillance threshold portended a faster time to first definitive treatment. CONCLUSION CCR outperforms other commonly used biomarkers for prediction of early metastasis. We illustrate the clinical utility of the CCR active surveillance and multimodality thresholds. Molecular genomic tests can inform patient selection and personalization of treatment for localized prostate cancer.
Collapse
Affiliation(s)
- Ryan J Hutten
- Department of Human Oncology, University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI
| | - Bismarck Odei
- Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah School of Medicine, Salt Lake City, UT
| | - Skyler B Johnson
- Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah School of Medicine, Salt Lake City, UT
| | - Jonathan D Tward
- Department of Radiation Oncology, Huntsman Cancer Hospital, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
8
|
Spratt DE, Liu VYT, Michalski J, Davicioni E, Berlin A, Simko JP, Efstathiou JA, Tran PT, Sandler HM, Hall WA, Thompson DJS, Parliament MB, Dayes IS, Correa RJM, Robertson JM, Gore EM, Doncals DE, Vigneault E, Souhami L, Karrison TG, Feng FY. Genomic Classifier Performance in Intermediate-Risk Prostate Cancer: Results From NRG Oncology/RTOG 0126 Randomized Phase 3 Trial. Int J Radiat Oncol Biol Phys 2023; 117:370-377. [PMID: 37137444 PMCID: PMC10949135 DOI: 10.1016/j.ijrobp.2023.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 03/15/2023] [Accepted: 04/12/2023] [Indexed: 05/05/2023]
Abstract
PURPOSE Intermediate-risk prostate cancer is a heterogeneous disease state with diverse treatment options. The 22-gene Decipher genomic classifier (GC) retrospectively has shown to improve risk stratification in these patients. We assessed the performance of the GC in men with intermediate-risk disease enrolled in NRG Oncology/RTOG 01-26 with updated follow-up. METHODS AND MATERIALS After National Cancer Institute approval, biopsy slides were collected from NRG Oncology/RTOG 01-26, a randomized phase 3 trial of men with intermediate-risk prostate cancer randomized to 70.2 Gy versus 79.2 Gy of radiation therapy without androgen deprivation therapy. RNA was extracted from the highest-grade tumor foci to generate the locked 22-gene GC model. The primary endpoint for this ancillary project was disease progression (composite of biochemical failure, local failure, distant metastasis, prostate cancer-specific mortality, and use of salvage therapy). Individual endpoints were also assessed. Fine-Gray or cause-specific Cox multivariable models were constructed adjusting for randomization arm and trial stratification factors. RESULTS Two-hundred fifteen patient samples passed quality control for analysis. The median follow-up was 12.8 years (range, 2.4-17.7). On multivariable analysis, the 22-gene GC (per 0.1 unit) was independently prognostic for disease progression (subdistribution hazard ratio [sHR], 1.12; 95% confidence interval [CI], 1.00-1.26; P = .04), biochemical failure (sHR, 1.22; 95% CI, 1.10-1.37; P < .001), distant metastasis (sHR, 1.28; 95% CI, 1.06-1.55; P = .01), and prostate cancer-specific mortality (sHR, 1.45; 95% CI, 1.20-1.76; P < .001). Ten-year distant metastasis in GC low-risk patients was 4% compared with 16% for GC high-risk patients. In patients with lower GC scores, the 10-year difference in metastasis-free survival rate between arms was -7%, compared with 21% for higher GC patients (P-interaction = .04). CONCLUSIONS This study represents the first validation of a biopsy-based gene expression classifier, assessing both its prognostic and predictive value, using data from a randomized phase 3 trial of intermediate-risk prostate cancer. Decipher improves risk stratification and can aid in treatment decision-making in men with intermediate-risk disease.
Collapse
Affiliation(s)
- Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Cleveland, Ohio.
| | | | - Jeff Michalski
- Department of Radiation Oncology, Washington University, St. Louis, Missouri
| | | | - Alejandro Berlin
- Princess Margaret Cancer Centre, Cancer Clinical Research Unit, Toronto, Ontario, Canada
| | - Jeffry P Simko
- Department of Pathology, UCSF Medical Center-Mount Zion, San Francisco, California
| | - Jason A Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Phuoc T Tran
- Department of Pathology, University of Maryland, Baltimore, Maryland
| | - Howard M Sandler
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California
| | - William A Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Matthew B Parliament
- Division of Radiation Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Ian S Dayes
- Division of Radiation Oncology, Hamilton Regional Cancer Centre, Ontario, Canada
| | | | - John M Robertson
- Department of Radiation Oncology, Beaumont Health CCOP, Royal Oak, Michigan
| | - Elizabeth M Gore
- Department of Radiation Oncology, Milwaukee VA Medical Center, Milwaukee, Wisconsin
| | | | - Eric Vigneault
- Department of Radiation Oncology, CHU de Quebec Universite Laval, Quebec, Canada
| | - Luis Souhami
- Department of Radiation Oncology, Cedars Cancer Centre, McGill University, Quebec, Canada
| | - Theodore G Karrison
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| |
Collapse
|
9
|
San Francisco IF, Rojas PA, Bravo JC, Díaz J, Ebel L, Urrutia S, Prieto B, Cerda-Infante J. Can We Predict Prostate Cancer Metastasis Based on Biomarkers? Where Are We Now? Int J Mol Sci 2023; 24:12508. [PMID: 37569883 PMCID: PMC10420177 DOI: 10.3390/ijms241512508] [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: 07/13/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The incidence of prostate cancer (PC) has risen annually. PC mortality is explained by the metastatic disease (mPC). There is an intermediate scenario in which patients have non-mPC but have initiated a metastatic cascade through epithelial-mesenchymal transition. There is indeed a need for more and better tools to predict which patients will progress in the future to non-localized clinical disease or already have micrometastatic disease and, therefore, will clinically progress after primary treatment. Biomarkers for the prediction of mPC are still under development; there are few studies and not much evidence of their usefulness. This review is focused on tissue-based genomic biomarkers (TBGB) for the prediction of metastatic disease. We develop four main research questions that we attempt to answer according to the current evidence. Why is it important to predict metastatic disease? Which tests are available to predict metastatic disease? What impact should there be on clinical guidelines and clinical practice in predicting metastatic disease? What are the current prostate cancer treatments? The importance of predicting metastasis is fundamental given that, once metastasis is diagnosed, quality of life (QoL) and survival drop dramatically. There is still a need and space for more cost-effective TBGB tests that predict mPC disease.
Collapse
Affiliation(s)
- Ignacio F. San Francisco
- Environ Innovation Laboratory, Avenida Providencia 1208 Oficina 207, Providencia, Santiago 7500000, Chile;
| | - Pablo A. Rojas
- Servicio de Urología, Complejo Asistencial Dr. Sotero del Río, Santiago 8150215, Chile;
| | - Juan C. Bravo
- Servicio de Urología, Hospital Regional Libertador Bernardo O’Higgins, Rancagua 2820000, Chile;
| | - Jorge Díaz
- Servicio de Urología, Instituto Oncológico Fundación Arturo López Pérez, Santiago 7500921, Chile;
| | - Luis Ebel
- Servicio de Urología, Hospital Base de Valdivia, Universidad Austral, Valdivia 5090000, Chile;
| | - Sebastián Urrutia
- Servicio de Urología, Hospital Dr. Hernán Henríquez Aravena, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Benjamín Prieto
- Environ Innovation Laboratory, Avenida Providencia 1208 Oficina 207, Providencia, Santiago 7500000, Chile;
| | - Javier Cerda-Infante
- Environ Innovation Laboratory, Avenida Providencia 1208 Oficina 207, Providencia, Santiago 7500000, Chile;
| |
Collapse
|
10
|
Herberts C, Wyatt AW, Nguyen PL, Cheng HH. Genetic and Genomic Testing for Prostate Cancer: Beyond DNA Repair. Am Soc Clin Oncol Educ Book 2023; 43:e390384. [PMID: 37207301 DOI: 10.1200/edbk_390384] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Significant progress has been made in genetic and genomic testing for prostate cancer across the disease spectrum. Molecular profiling is increasingly relevant for routine clinical management, fueled in part by advancements in testing technology and integration of biomarkers into clinical trials. In metastatic prostate cancer, defects in DNA damage response genes are now established predictors of benefit to US Food and Drug Administration-approved poly (ADP-ribose) polymerase inhibitors and immune checkpoint inhibitors, and trials are actively investigating these and other targeted treatment strategies in earlier disease states. Excitingly, opportunities for molecularly informed management beyond DNA damage response genes are also maturing. Germline genetic variants (eg, BRCA2 or MSH2/6) and polygenic germline risk scores are being investigated to inform cancer screening and active surveillance in at-risk carriers. RNA expression tests have recently gained traction in localized prostate cancer, enabling patient risk stratification and tailored treatment intensification via radiotherapy and/or androgen deprivation therapy for localized or salvage treatment. Finally, emerging minimally invasive circulating tumor DNA technology promises to enhance biomarker testing in advanced disease pending additional methodological and clinical validation. Collectively, genetic and genomic tests are rapidly becoming indispensable tools for informing the optimal clinical management of prostate cancer.
Collapse
Affiliation(s)
- Cameron Herberts
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Paul L Nguyen
- Harvard Medical School, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA
| | - Heather H Cheng
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
11
|
In Regard to Dess. Int J Radiat Oncol Biol Phys 2023; 115:261-263. [DOI: 10.1016/j.ijrobp.2022.08.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/22/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022]
|
12
|
Spohn SKB, Draulans C, Kishan AU, Spratt D, Ross A, Maurer T, Tilki D, Berlin A, Blanchard P, Collins S, Bronsert P, Chen R, Pra AD, de Meerleer G, Eade T, Haustermans K, Hölscher T, Höcht S, Ghadjar P, Davicioni E, Heck M, Kerkmeijer LGW, Kirste S, Tselis N, Tran PT, Pinkawa M, Pommier P, Deltas C, Schmidt-Hegemann NS, Wiegel T, Zilli T, Tree AC, Qiu X, Murthy V, Epstein JI, Graztke C, Gao X, Grosu AL, Kamran SC, Zamboglou C. Genomic Classifiers in Personalized Prostate Cancer Radiation Therapy Approaches: A Systematic Review and Future Perspectives Based on International Consensus. Int J Radiat Oncol Biol Phys 2022:S0360-3016(22)03691-4. [PMID: 36596346 DOI: 10.1016/j.ijrobp.2022.12.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/09/2022] [Accepted: 12/24/2022] [Indexed: 01/01/2023]
Abstract
Current risk-stratification systems for prostate cancer (PCa) do not sufficiently reflect the disease heterogeneity. Genomic classifiers (GC) enable improved risk stratification after surgery, but less data exist for patients treated with definitive radiation therapy (RT) or RT in oligo-/metastatic disease stages. To guide future perspectives of GCs for RT, we conducted (1) a systematic review on the evidence of GCs for patients treated with RT and (2) a survey of experts using the Delphi method, addressing the role of GCs in personalized treatments to identify relevant fields of future clinical and translational research. We performed a systematic review and screened ongoing clinical trials on ClinicalTrials.gov. Based on these results, a multidisciplinary international team of experts received an adapted Delphi method survey. Thirty-one and 30 experts answered round 1 and round 2, respectively. Questions with ≥75% agreement were considered relevant and included in the qualitative synthesis. Evidence for GCs as predictive biomarkers is mainly available to the postoperative RT setting. Validation of GCs as prognostic markers in the definitive RT setting is emerging. Experts used GCs in patients with PCa with extensive metastases (30%), in postoperative settings (27%), and in newly diagnosed PCa (23%). Forty-seven percent of experts do not currently use GCs in clinical practice. Expert consensus demonstrates that GCs are promising tools to improve risk-stratification in primary and oligo-/metastatic patients in addition to existing classifications. Experts were convinced that GCs might guide treatment decisions in terms of RT-field definition and intensification/deintensification in various disease stages. This work confirms the value of GCs and the promising evidence of GC utility in the setting of RT. Additional studies of GCs as prognostic biomarkers are anticipated and form the basis for future studies addressing predictive capabilities of GCs to optimize RT and systemic therapy. The expert consensus points out future directions for GC research in the management of PCa.
Collapse
Affiliation(s)
- Simon K B Spohn
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Cédric Draulans
- Department of Radiation Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU Leuven, Belgium
| | - Amar U Kishan
- Departments of Radiation Oncology and Urology, University of California, Los Angeles, California
| | - Daniel Spratt
- Department of Radiation Oncology, UH Seidman Cancer Center, Case Western Reserve University
| | - Ashley Ross
- Department of Urology, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Alejandro Berlin
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, and Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network. Toronto, Canada
| | - Pierre Blanchard
- Department of Radiation Oncology, Gustave Roussy, Oncostat U1018, Inserm, Paris-Saclay University, Villejuif, France
| | - Sean Collins
- Department of Radiation Medicine, Medstar Georgetown University Hospital, Washington, DC
| | - Peter Bronsert
- Institute for Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ronald Chen
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, Kansas
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami, Miller School of Medicine
| | - Gert de Meerleer
- Department of Radiation Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU Leuven, Belgium
| | - Thomas Eade
- Northern Sydney Cancer Centre, Radiation Oncology Unit, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Karin Haustermans
- Department of Radiation Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU Leuven, Belgium
| | - Tobias Hölscher
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan Höcht
- Xcare Practices Dept. Radiotherapy, Saarlouis, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin
| | | | - Matthias Heck
- Department of Urology, Rechts der Isar Medical Center, Technical University of Munich, Germany
| | - Linda G W Kerkmeijer
- Department of Radiation Oncology, Radboud University Medical Center, The Netherlands
| | - Simon Kirste
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Nikolaos Tselis
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland
| | - Michael Pinkawa
- Department of Radiation Oncology, MediClin Robert Janker Klinik Bonn, Germany
| | - Pascal Pommier
- Department of Radiation Oncology, Centre Léon Bérard, Lyon, France
| | - Constantinos Deltas
- Molecular Medicine Research Center and Laboratory of Molecular and Medical Genetics, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | | | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Thomas Zilli
- Department of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Alison C Tree
- Department of Radiotherapy, Royal Marsden Hospital and the Institute of Cancer Research, London, United Kingdom
| | - Xuefeng Qiu
- Department of Urology, Medical School of Nanjing University, Affiliated Drum Tower Hospital, Nanjing, China
| | - Vedang Murthy
- Department of Radiation Oncology, ACTREC, Tata Memorial Centre, Homi Bhabha National University, India
| | - Jonathan I Epstein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christian Graztke
- Department of Urology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xin Gao
- Department of Internal Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Anca L Grosu
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Constantinos Zamboglou
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Oncology Center, European University of Cyprus, Limassol, Cyprus
| |
Collapse
|
13
|
Fuller DB, Crabtree T, Kane BL, Medbery CA, Pfeffer R, Gray JR, Peddada A, Royce TJ, Chen RC. High Dose “HDR-Like” Prostate SBRT: PSA 10-Year Results From a Mature, Multi-Institutional Clinical Trial. Front Oncol 2022; 12:935310. [PMID: 35965547 PMCID: PMC9373838 DOI: 10.3389/fonc.2022.935310] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose/Objective(s) Although ample intermediate-term prostate stereotactic body radiotherapy (SBRT) outcomes have been reported, 10-year results remain relatively sparse. Materials/Methods Eighteen institutions enrolled 259 low- and intermediate-risk patients. Median follow-up is 5.5 years, with 66 patients followed ≥ 10 years. This SBRT regimen specifically emulated an existing HDR brachytherapy dose schedule and isodose morphology, prescribed to 38 Gy/4 fractions, delivered daily by robotic SBRT, mandating > 150% dose escalation in the peripheral zone. Androgen deprivation therapy was not allowed, and a hydrogel spacer was not available at that time. Results Median pre-SBRT PSA 5.12 ng/mL decreased to 0.1 ng/mL by 3.5 years, with further decrease to a nadir of < 0.1 ng/mL by 7 years, maintained through 10 years. Ten-year freedom from biochemical recurrence measured 100% for low-risk, 84.3% for favorable intermediate risk (FIR), and 68.4% for unfavorable intermediate (UIR) cases. Multivariable analysis revealed that the UIR group bifurcated into two distinct prognostic subgroups. Those so classified by having Gleason score 4 + 3 and/or clinical stage T2 (versus T1b/T1c) had a significantly poorer 10 year freedom from biochemical recurrence rate, 54.8% if either or both factors were present, while UIR patients without these specific factors had a 94.4% 10-year freedom from biochemical recurrence rate. The cumulative incidence of grade 2 GU toxicity modestly increased over time – 16.3% at 5 years increased to 19.2% at 10 years-- while the incidence of grade 3+ GU and GI toxicity remained low and stable to 10 years - 2.6% and 0%, respectively. The grade 2 GI toxicity incidence also remained low and stable to 10 years – 4.1% with no further events after year 5. Conclusion This HDR-like SBRT regimen prescribing 38 Gy/4 fractions but delivering much higher intraprostatic doses on a daily basis is safe and effective. This treatment achieves a median PSA nadir of <0.1 ng/mL and provides high long-term disease control rates without ADT except for a subgroup of unfavorable intermediate-risk patients.
Collapse
Affiliation(s)
- Donald B. Fuller
- CyberKnife Centers of San Diego, San Diego, CA, United States
- *Correspondence: Donald B. Fuller, ; Ronald C. Chen,
| | - Tami Crabtree
- Advance Research Associates, Santa Clara, CA, United States
| | | | | | - Robert Pfeffer
- Benefis Sletten Cancer Institute, Great Falls, MT, United States
| | - James R. Gray
- Sarah Cannon Research Institute, Nashville, TN, United States
| | - Anuj Peddada
- Penrose-St. Francis Health Services, Colorado Springs, CO, United States
| | - Trevor J. Royce
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ronald C. Chen
- University of Kansas, Kansas City, KS, United States
- *Correspondence: Donald B. Fuller, ; Ronald C. Chen,
| |
Collapse
|
14
|
Kuhl V, Clegg W, Meek S, Lenz L, Flake DD, Ronan T, Kornilov M, Horsch D, Scheer M, Farber D, Zalaznick H, Cussenot O, Compérat E, Cancel-Tassin G, Wild PJ, Chun FK, Mandel P, Moinfar F, Cohen T, Delee S, Kronenwett R, Doedt J. Development and validation of a cell cycle progression signature for decentralized testing of men with prostate cancer. Biomark Med 2022; 16:449-459. [PMID: 35321552 DOI: 10.2217/bmm-2021-0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: The 46-gene Prolaris® cell cycle progression test provides information on the risk of prostate cancer progression. Here we developed and validated a 16-gene kit-based version. Methods: RNA was extracted from prostate cancer biopsy tissue. Amplification efficiency, minimum tumor content, repeatability, reproducibility and equivalence with the 46-gene test were evaluated. Results: Amplification efficiencies for all genes were within the acceptable range (90-110%), and samples with ≥50% tumor content were appropriate for the 16-gene test. Results were repeatable (standard deviation: 0.085) and reproducible (standard deviation: 0.115). Instrument, operator and kit lot had minimal impact on results. Cell cycle progression scores from the 46- and 16-gene tests were highly correlated (r = 0.969; bias = 0.217). Conclusion: The 16-gene test performs consistently and similarly to the 46-gene test.
Collapse
Affiliation(s)
- Vanessa Kuhl
- Myriad International GmbH, Cologne, 50829, Germany
| | - Wyatt Clegg
- Myriad Genetics, Inc., Salt Lake City, UT 84108, USA
| | | | - Lauren Lenz
- Myriad Genetics, Inc., Salt Lake City, UT 84108, USA
| | - Darl D Flake
- Myriad Genetics, Inc., Salt Lake City, UT 84108, USA
| | - Tracy Ronan
- Myriad Genetics, Inc., Salt Lake City, UT 84108, USA
| | - Max Kornilov
- Myriad International GmbH, Cologne, 50829, Germany
| | | | | | | | | | | | - Eva Compérat
- Department of Pathology, Sorbonne University, Tenon Hospital, Paris, 75020, France
| | | | - Peter J Wild
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, 60596, Germany.,WILDLAB, University Hospital Frankfurt MVZ GmbH, Frankfurt am Main, 60596, Germany
| | - Felix Kh Chun
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, 60596, Germany
| | - Philipp Mandel
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, 60596, Germany
| | - Farid Moinfar
- Department of Pathology, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, 4010, Austria
| | - Todd Cohen
- Myriad Genetics, Inc., Salt Lake City, UT 84108, USA
| | | | | | | |
Collapse
|