1
|
Castaneda P, Kuhlmann PK, Ithisuphalap J, Howard LE, Klaassen Z, Rivera LG, Amling CL, Aronson WJ, Cooperberg MR, Kane CJ, Terris MK, Freedland SJ. Alternate definitions of adverse pathology to predict a very high risk of metastasis in men with intermediate- and low-risk prostate cancer. Cancer 2025; 131:e35684. [PMID: 39748487 DOI: 10.1002/cncr.35684] [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: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Adverse pathology (AP) is often used as an intermediate end point for long-term outcomes in men with prostate cancer (PCa) who are active surveillance candidates. The association between a commonly used AP definition and long-term outcomes was tested, which identified definitions more strongly linked to a high risk of metastasis. METHODS Data were reviewed from the Shared Equal Access Regional Cancer Hospital cohort of men undergoing radical prostatectomy (RP) from 1988 to 2020 at nine Veterans Affairs hospitals. Men meeting National Comprehensive Cancer Network low-risk and favorable intermediate-risk criteria were included. Men with and without AP were compared; men with AP were defined as having grade groups 3-5 or pathological stage ≥pT3a or pN1 at RP (definition 1). Sensitivity analyses were performed for six alternative definitions (definitions 2-7) and their association with biochemical recurrence (BCR), metastasis, PCa-specific mortality (PCSM), and castrate-resistant PCa (CRPC). RESULTS A total of 2175 men were included: 711 had AP by definition 1. In univariable analyses, all AP definitions were associated with the risk of BCR, metastasis, and PCSM. All but one definition were associated with CRPC. In definitions 1-6, the 10-year event rate for metastasis in those with AP ranged from 3.0% (definition 1) to 7.9% (definition 5). Only in definition 7 was the 10-year event rate for metastasis >10%. However, only 0.5% of patients (11 of 2175) met definition 7. CONCLUSIONS AP was statistically associated with relatively worse outcomes. However, in all but the most stringent definitions, met by <1% of patients, the absolute event rate of metastasis in men with AP was low. This challenges the clinical usefulness of AP as an intermediate end point in men with intermediate- to low-risk PCa.
Collapse
Affiliation(s)
- Peris Castaneda
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Paige K Kuhlmann
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jaruda Ithisuphalap
- Division of Urology, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | - Lauren E Howard
- Division of Urology, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | - Zachary Klaassen
- Department of Urology, Augusta University, Augusta, Georgia, USA
| | - Lourdes Guerrios Rivera
- Urology Section, Surgery Department, Veterans Administration Caribbean Health Care System, San Juan, Puerto Rico
| | - Christopher L Amling
- Department of Urology, Oregon Health and Science University, Portland, Oregon, USA
| | - William J Aronson
- Department of Urology, University of California Los Angeles Medical Center, Los Angeles, California, USA
| | - Matthew R Cooperberg
- Department of Urology, University of California San Francisco Medical Center, San Francisco, California, USA
| | - Christopher J Kane
- Department of Urology, University of California San Diego Health System, San Diego, California, USA
| | - Martha K Terris
- Department of Urology, Augusta University, Augusta, Georgia, USA
| | - Stephen J Freedland
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Division of Urology, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| |
Collapse
|
2
|
Quan Y, Wang M, Zhang H, Lu D, Ping H. Spatial transcriptomics identifies RBM39 as a gene a ssociated with Gleason score progression in prostate cancer. iScience 2024; 27:111351. [PMID: 39650727 PMCID: PMC11625293 DOI: 10.1016/j.isci.2024.111351] [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: 06/11/2024] [Revised: 09/17/2024] [Accepted: 11/06/2024] [Indexed: 12/11/2024] Open
Abstract
Prostate cancer (PCa) exhibits significant intratumor heterogeneity, frequently manifesting as a multifocal disease. This study utilized Visium spatial transcriptomics (ST) to explore transcriptome patterns in PCa regions with varying Gleason scores (GSs). Principal component analysis (PCA) and Louvain clustering analysis revealed transcriptomic classifications aligned with the histology of different GSs. The increasing degree of tumor malignancy during GS progression was validated using inferred copy number variation (inferCNV) analysis. Diffusion pseudotime (DPT) and partition-based graph abstraction (PAGA) analyses predicted the developmental trajectories among distinct clusters. Differentially expressed gene (DEG) analysis through pairwise comparisons of various GSs identified genes associated with GS progression. Validation with The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) dataset confirmed the differential expression of RBM39, a finding further supported by cytological and histological experiments. These findings enhance our understanding of GS evolution through spatial transcriptomics and highlight RBM39 as a gene associated with GS progression.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, P.R. China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, P.R. China
| | - Hong Zhang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, P.R. China
| | - Dan Lu
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing 100191, P.R. China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, P.R. China
| |
Collapse
|
3
|
Quan Y, Zhang H, Wang M, Ping H. UQCRB and LBH are correlated with Gleason score progression in prostate cancer: Spatial transcriptomics and experimental validation. Comput Struct Biotechnol J 2024; 23:3315-3326. [PMID: 39310280 PMCID: PMC11414276 DOI: 10.1016/j.csbj.2024.08.026] [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: 06/08/2024] [Revised: 08/09/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Prostate cancer (PCa) is a multifocal disease characterized by genomic and phenotypic heterogeneity within a single gland. In this study, Visium spatial transcriptomics (ST) analysis was applied to PCa tissues with different histological structures to infer the molecular events involved in Gleason score (GS) progression. The spots in tissue sections were classified into various groups using Principal Component Analysis (PCA) and Louvain clustering analysis based on transcriptome data. Anotation of the spots according to GS revealed notable similarities between transcriptomic profiles and histologically identifiable structures. The accuracy of macroscopic GS determination was bioinformatically verified through malignancy-related feature analysis, specifically inferred copy number variation (inferCNV), as well as developmental trajectory analyses, such as diffusion pseudotime (DPT) and partition-based graph abstraction (PAGA). Genes related to GS progression were identified from the differentially expressed genes (DEGs) through pairwise comparisons of groups along a GS gradient. The proteins encoded by the representative oncogenes UQCRB and LBH were found to be highly expressed in advanced-stage PCa tissues. Knockdown of their mRNAs significantly suppressed PCa cell proliferation and invasion. These findings were validated using The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) dataset, as well as through histological and cytological experiments. The results presented here establish a foundation for ST-based evaluation of GS progression and provide valuable insights into the GS progression-related genes UQCRB and LBH.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, China
| | - Hong Zhang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100176, China
| |
Collapse
|
4
|
Sood A, Grauer R, Diaz-Insua M, Tewari AK, Hemal AK, Shrivastava A, Peabody JO, Jeong W, Abdollah F, Rudzinski JK, Andrews JR, Gorin MA, Bhandari M, Menon M. 15-year biochemical failure, metastasis, salvage therapy, and cancer-specific and overall survival rates in men treated with robotic radical prostatectomy for PSA-screen detected prostate cancer. Prostate Cancer Prostatic Dis 2023; 26:778-786. [PMID: 37142635 DOI: 10.1038/s41391-023-00674-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND An informed decision regarding a treatment option requires data on its long-term efficacy and side-effect profile. While the side-effects of robotic radical prostatectomy have been well-quantified, the data on its long-term efficacy are lacking. We here provide 15-year oncological outcomes of clinically-localized prostate cancer (CLPCa) patients treated with robot-assisted laparoscopic prostatectomy (RALP). METHODS We treated 1,807 men with CLPCa with RALP between 2001 and 2005 and prospectively collected follow-up data through 2020. We examined the rates of biochemical failure (BCF), metastatic progression, secondary therapy use, PCa-specific mortality (PCSM), and overall survival (OS) using Kaplan-Meier and competing-risk cumulative incidence methods as appropriate. RESULTS The median follow-up was 14.1 years. Six hundred eight and 312 men had D'Amico intermediate- and high-risk disease, respectively. Overall, the 15-year rates of BCF, metastasis, secondary therapy use, PCSM, and OS were 28.1%, 4.0%, 16.3%, 2.5%, and 82.1%, respectively. The rates of oncologic failure increased with increasing D'Amico (preoperative) and Diaz (postoperative) risk scores - BCF, metastasis, and PCSM rates in D'Amico low-, intermediate-, and high-risk groups at 15-years were 15.2%, 38.3%, and 44.1% [BCF], 1.1%, 4.1%, and 13.0% [metastasis], and 0.5%, 3.4%, and 6.6% [PCSM], respectively, and in Diaz risk groups 1, 2, 3, 4, and 5 were 5.5%, 20.6%, 41.8%, 66.9%, and 89.2% [BCF], 0%, 0.5%, 3.2%, 20.5%, and 60.0% [metastasis], and 0%, 0.8%, 0.6%, 13.5%, and 37.5% [PCSM], respectively. The OS rates in D'Amico low-to-high and Diaz 1-to-5 risk groups at 15-years were 85.9%, 78.6%, and 75.2%, and 89.4%, 83.2%, 80.6%, 67.2%, and 23.4%, respectively. CONCLUSIONS Men diagnosed with clinically-localized prostate cancer in the contemporaneous PSA-screening era and treated with RALP achieve durable long-term oncological control. The data reported here (in a risk-stratified manner) represent the longest follow-up after robotic radical prostatectomy, and as such, should be of value when counseling patients regarding expected oncologic outcomes from RALP.
Collapse
Affiliation(s)
- Akshay Sood
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA.
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA.
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Urology, The James Cancer Hospital and Solove Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Ralph Grauer
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mireya Diaz-Insua
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashok K Hemal
- Department of Urology, Wake Forest University, Winston-Salem, NC, USA
| | | | - James O Peabody
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Wooju Jeong
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Firas Abdollah
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Jan K Rudzinski
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack R Andrews
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Gorin
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Mani Menon
- VCORE - Vattikuti Urology Institute Center for Outcomes Research, Analytics and Evaluation, Henry Ford Hospital, Detroit, MI, USA
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
5
|
Zenner ML, Kirkpatrick B, Leonardo TR, Schlicht MJ, Saldana AC, Loitz C, Valyi‐Nagy K, Maienschein‐Cline M, Gann PH, Abern M, Nonn L. Prostate-derived circulating microRNAs add prognostic value to prostate cancer risk calculators. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e122. [PMID: 38496750 PMCID: PMC10938556 DOI: 10.1002/jex2.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/18/2023] [Accepted: 10/11/2023] [Indexed: 03/19/2024]
Abstract
Prostate cancer is the second leading cause of malignancy-related deaths among American men. Active surveillance is a safe option for many men with less aggressive disease, yet definitively determining low-risk cancer is challenging with biopsy alone. Herein, we sought to identify prostate-derived microRNAs in patient sera and serum extracellular vesicles, and determine if those microRNAs improve upon the current clinical risk calculators for prostate cancer prognosis before and after biopsy. Prostate-derived intracellular and extracellular vesicle-contained microRNAs were identified by small RNA sequencing of prostate cancer patient explants and primary cells. Abundant microRNAs were included in a custom microRNA PCR panel that was queried in whole serum and serum extracellular vesicles from a diverse cohort of men diagnosed with prostate cancer. The levels of these circulating microRNAs significantly differed between indolent and aggressive disease and improved the area under the curve for pretreatment nomograms of prostate cancer disease risk. The microRNAs within the extracellular vesicles were the most informative and improved the AUC to 0.739 compared to the existing nomogram alone, which has an AUC of 0.561. The microRNAs in the whole serum improved it to AUC 0.675. In summary, quantifying microRNAs circulating in extracellular vesicles is a clinically feasible assay that may provide additional information for assessing prostate cancer risk stratification.
Collapse
Affiliation(s)
- Morgan L. Zenner
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- University of Illinois Cancer CenterChicagoIllinoisUSA
| | - Brenna Kirkpatrick
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- University of Illinois Cancer CenterChicagoIllinoisUSA
| | - Trevor R. Leonardo
- Department of Microbiology and ImmunologyUniversity of Illinois ChicagoChicagoIllinoisUSA
- Department of Periodontics, Center for Wound Healing and Tissue RegenerationUniversity of Illinois ChicagoChicagoIllinoisUSA
| | | | - Alejandra Cavazos Saldana
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- University of Illinois Cancer CenterChicagoIllinoisUSA
| | - Candice Loitz
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- University of Illinois Cancer CenterChicagoIllinoisUSA
| | - Klara Valyi‐Nagy
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Mark Maienschein‐Cline
- Research Resource Core BioinformaticsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Peter H. Gann
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- University of Illinois Cancer CenterChicagoIllinoisUSA
| | - Michael Abern
- Department of UrologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Larisa Nonn
- Department of PathologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- University of Illinois Cancer CenterChicagoIllinoisUSA
| |
Collapse
|
6
|
Quan Y, Ping H, Wang M, Zhang X. RNA-Sequencing Analysis Indicates That N-Cadherin Promotes Prostate Cancer Progression by the Epigenetic Modification of Key Genes. DNA Cell Biol 2023; 42:563-577. [PMID: 37540080 DOI: 10.1089/dna.2023.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023] Open
Abstract
N-cadherin (cadherin-2 [CDH2]) is widely known as the promoter of prostate cancer (PCa) invasion and castration resistance. However, the biological mechanism of N-cadherin in PCa progression is unclear. In this study, we overexpressed N-cadherin in LNCaP cells and downregulated N-cadherin in PC3 cells by lentiviral transduction. Then, differentially expressed genes (DEGs) and dysregulated biological functions were investigated through RNA sequencing (RNA-seq) analyses. We found 13 long noncoding RNA (lncRNA) transcripts, 72 messenger RNA (mRNA) transcripts, and 3 integrated genes were dysregulated by N-cadherin. In the disease enrichment, bone cancer, and neurodegenerative and nervous system diseases were associated with N-cadherin in the circular RNA (circRNA; PC3 versus [vs.,/] LNCaP [PC3/LNCaP] comparison) and DEG analysis (LNCaP_oe_CDH2 vs. LNCaP_oe_NC [LNCaP_oe_CDH2/NC] comparison). Epigenetic reprogramming, such as nucleic acid binding, and chromatin and histone modifications, was enriched in Gene Ontology (GO) analysis (DEGs in LNCaP_oe_CDH2/NC and PC3_sh_NC/CDH2, and host genes of circRNA in PC3/LNCaP). Transcriptional misregulation in cancer, post-translational protein modification, gene expression, and generic transcription pathways were dysregulated in the pathway enrichment analysis (host genes of circRNA in PC3/LNCaP, and DEGs in LNCaP_oe_CDH2/NC and PC3_sh_NC/CDH2). Verifying DEGs through TCGA-PRAD dataset revealed six oncogenes (ARHGEF1, GRAMD1A, GTF2H4, MAPK8IP3, POLD1, and PTBP1) that were commonly upregulated by N-cadherin and in advanced PCa stages. In summary, we identified several oncogenes and biological functions associated with N-cadherin expression in PCa cells. N-cadherin may trigger epigenetic reprogramming in PCa cells to promote tumor progression.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Quan Y, Zhang X, Wang M, Ping H. Histone lysine methylation patterns in prostate cancer microenvironment infiltration: Integrated bioinformatic analysis and histological validation. Front Oncol 2022; 12:981226. [PMID: 36237332 PMCID: PMC9552767 DOI: 10.3389/fonc.2022.981226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
Background Epigenetic reprogramming through dysregulated histone lysine methylation (HLM) plays a crucial role in prostate cancer (PCa) progression. This study aimed to comprehensively evaluate HLM modification patterns in PCa microenvironment infiltration. Materials and methods Ninety-one HLM regulators in The Cancer Genome Atlas (TCGA) dataset were analyzed using bioinformatics. Differentially expressed genes (DEGs) and survival analyses were performed using TCGA-PRAD clinicopathologic and follow-up information. Consensus clustering analysis divided patients into subgroups. Gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the DEGs. Tumor mutation burden (TMB) and tumor microenvironment (TME) cell infiltration were evaluated in different HLM clusters. Quantitative real-time PCR (qPCR) analysis assessed HLM regulators in clinical PCa tissues. Results The tumor vs. normal (TN), Gleason score (GS) > 7 vs. GS < 7, pathological T stage (pT) = 2 vs. pT = 3, and TP53 mutation vs. wild-type comparisons using TCGA-PRAD dataset revealed 3 intersecting HLM regulators (EZH2, NSD2, and KMT5C) that were consistently upregulated in advanced PCa (GS > 7, pT3, HR > 1, and TP53 mutation) (P < 0.05) and verified in clinical PCa tissues. Consensus clustering analysis revealed three distinct HLM modification patterns (HLMclusters). However, no significant differences in recurrence-free survival (RFS) rates were found among the groups (P > 0.05). We screened 189 HLM phenotype-related genes that overlapped in the pairwise comparisons of HLMclusters and P < 0.01 in the Cox regression analysis. Three distinct subgroups (geneClusters) were revealed based on the 189 genes, in which cluster A involved the most advanced PCa (PSA > 10, T3-4, GS8-10, and biochemical recurrence) and the poorest RFS. The HLM score (HLMscore) was calculated by principal component analysis (PCA) of HLM phenotype-related genes that have positive predictive value for RFS (P < 0.001) and immune therapy responses (in the CTLA4-positive and -negative responses accompanied by a PD1-negative response). Conclusion We comprehensively evaluated HLM regulators in the PCa microenvironment using TCGA-PRAD, revealing a nonnegligible role of HLM patterns in PCa complexity and heterogeneity. Elucidating the effects of HLM regulators in PCa may enhance prognostics, aggressiveness assessments, and immunotherapy strategies.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing Tongren Hospital, Beijing, China
- *Correspondence: Hao Ping,
| |
Collapse
|
8
|
Paulson N, Zeevi T, Papademetris M, Leapman MS, Onofrey JA, Sprenkle PC, Humphrey PA, Staib LH, Levi AW. Prediction of Adverse Pathology at Radical Prostatectomy in Grade Group 2 and 3 Prostate Biopsies Using Machine Learning. JCO Clin Cancer Inform 2022; 6:e2200016. [PMID: 36179281 DOI: 10.1200/cci.22.00016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE There is ongoing clinical need to improve estimates of disease outcome in prostate cancer. Machine learning (ML) approaches to pathologic diagnosis and prognosis are a promising and increasingly used strategy. In this study, we use an ML algorithm for prediction of adverse outcomes at radical prostatectomy (RP) using whole-slide images (WSIs) of prostate biopsies with Grade Group (GG) 2 or 3 disease. METHODS We performed a retrospective review of prostate biopsies collected at our institution which had corresponding RP, GG 2 or 3 disease one or more cores, and no biopsies with higher than GG 3 disease. A hematoxylin and eosin-stained core needle biopsy from each site with GG 2 or 3 disease was scanned and used as the sole input for the algorithm. The ML pipeline had three phases: image preprocessing, feature extraction, and adverse outcome prediction. First, patches were extracted from each biopsy scan. Subsequently, the pre-trained Visual Geometry Group-16 convolutional neural network was used for feature extraction. A representative feature vector was then used as input to an Extreme Gradient Boosting classifier for predicting the binary adverse outcome. We subsequently assessed patient clinical risk using CAPRA score for comparison with the ML pipeline results. RESULTS The data set included 361 WSIs from 107 patients (56 with adverse pathology at RP). The area under the receiver operating characteristic curves for the ML classification were 0.72 (95% CI, 0.62 to 0.81), 0.65 (95% CI, 0.53 to 0.79) and 0.89 (95% CI, 0.79 to 1.00) for the entire cohort, and GG 2 and GG 3 patients, respectively, similar to the performance of the CAPRA clinical risk assessment. CONCLUSION We provide evidence for the potential of ML algorithms to use WSIs of needle core prostate biopsies to estimate clinically relevant prostate cancer outcomes.
Collapse
Affiliation(s)
| | - Tal Zeevi
- Yale School of Medicine, New Haven, CT
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Morote J, Borque-Fernando A, Triquell M, Celma A, Regis L, Mast R, de Torres IM, Semidey ME, Santamaría A, Planas J, Esteban LM, Trilla E. Multiparametric Magnetic Resonance Imaging Grades the Aggressiveness of Prostate Cancer. Cancers (Basel) 2022; 14:1828. [PMID: 35406600 PMCID: PMC8997549 DOI: 10.3390/cancers14071828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
We sought to find further evidence showing the increase in PCa aggressiveness as PI-RADS score increases from four surrogates of PCa aggressiveness: i. prostate biopsy GG (≤3 vs. >3), ii. type of pathology in surgical specimens (favourable vs. unfavourable), iii. clinical stage (localised vs. advanced), and risk of recurrence of localised PCa after primary treatment (low-intermediate vs. high). A group of 692 PCa patients were diagnosed after 3-T multiparametric MRI (mpMRI) and guided and/or systematic biopsies, showing csPCa (GG ≥ 2) in 547 patients (79%) and insignificant PCa (iPCa) in 145 (21%). The csPCa rate increased from 32.4% in PI-RADS < 3 to 95.5% in PI-RADS 5 (p < 0.001). GG ≥ 3 was observed in 7.6% of PCa with PI-RADS < 3 and 32.6% in those with PI-RADS > 3 (p < 0.001). Unfavourable pathology was observed in 38.9% of PCa with PI-RAD < 3 and 68.3% in those with PI-RADS > 3 (p = 0.030). Advanced disease was not observed in PCa with PI-RADS ≤ 3, while it existed in 12.7% of those with PI-RADS > 3 (p < 0.001). High-risk recurrence localised PCa was observed in 9.5% of PCa with PI-RADS < 3 and 35% in those with PI-RADS > 3 (p = 0.001). The PI-RADS score was an independent predictor of all surrogates of PCa aggressiveness as PSA density. We confirmed that mpMRI grades PCa aggressiveness.
Collapse
Affiliation(s)
- Juan Morote
- Department of Urology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (M.T.); (A.C.); (L.R.); (J.P.); (E.T.)
- Department of Surgery, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Angel Borque-Fernando
- Department of Urology, Hospital Universitario Miguel Servet, IIS-Aragon, 50009 Zaragoza, Spain;
| | - Marina Triquell
- Department of Urology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (M.T.); (A.C.); (L.R.); (J.P.); (E.T.)
| | - Anna Celma
- Department of Urology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (M.T.); (A.C.); (L.R.); (J.P.); (E.T.)
| | - Lucas Regis
- Department of Urology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (M.T.); (A.C.); (L.R.); (J.P.); (E.T.)
| | - Richard Mast
- Department of Radiology, Vall d’Hebron Hospital, 08035 Barcelona, Spain;
| | - Inés M. de Torres
- Department of Pathology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (I.M.d.T.); (M.E.S.)
- Department of Morphological Sciences, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - María E. Semidey
- Department of Pathology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (I.M.d.T.); (M.E.S.)
- Department of Morphological Sciences, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | | | - Jacques Planas
- Department of Urology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (M.T.); (A.C.); (L.R.); (J.P.); (E.T.)
| | - Luis M. Esteban
- Department of Applied Mathematics, Escuela Universitaria Politécnica La Almunia, Universidad de Zaragoza, 50100 Zaragoza, Spain;
| | - Enrique Trilla
- Department of Urology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (M.T.); (A.C.); (L.R.); (J.P.); (E.T.)
- Department of Surgery, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
10
|
Athanasiou A, Tennstedt P, Wittig A, Huber R, Straub O, Schiess R, Steuber T. A novel serum biomarker quintet reveals added prognostic value when combined with standard clinical parameters in prostate cancer patients by predicting biochemical recurrence and adverse pathology. PLoS One 2021; 16:e0259093. [PMID: 34767586 PMCID: PMC8589165 DOI: 10.1371/journal.pone.0259093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/12/2021] [Indexed: 11/18/2022] Open
Abstract
The objective was to determine the prognostic utility of a new biomarker combination in prostate cancer (PCa) patients undergoing Radical Prostatectomy (RP). Serum samples and clinical data of 557 men who underwent RP for PCa with pathological stage (pT) <3 at Martini Clinic (Hamburg, Germany) were used for analysis. Clinical Grade Group and clinical stage was determined using biopsy samples while tumor marker concentrations were measured in serum using immunoassays. The prognostic utility of the proposed marker combination was assessed using Cox proportional hazard regression and Kaplan-Meier analysis. The performance was compared to the Cancer of the Prostate Risk Assessment (CAPRA) score in the overall cohort and in a low-risk patient subset. A multivariable model comprising fibronectin 1, galectin-3-binding protein, lumican, matrix metalloprotease 9, thrombospondin-1 and PSA together with clinical Grade Group (GG) and clinical stage (cT) was created. The proposed model was a significant predictor of biochemical recurrence (BCR) (HR 1.29 per 5 units score, 95%CI 1.20–1.38, p<0.001). The Kaplan-Meier analysis showed that the proposed model had a better prediction for low-risk disease after RP compared to CAPRA (respectively 5.0% vs. 9.1% chance of BCR). In a pre-defined low risk population subset, the risk of BCR using the proposed model was below 5.2% and thus lower when compared to CAPRA = 0–2 (9%), GG<2 (7%) and NCCN = low-risk (6%) subsets. Additionally, the proposed model could significantly (p<0.001) discriminate patients with adverse pathology (AP) events at RP from those without. In conclusion, the proposed model is superior to CAPRA for the prediction of BCR after RP in the overall cohort as well as a in a pre-defined low risk patient population subset. It is also significantly associated with AP at RP.
Collapse
Affiliation(s)
| | - Pierre Tennstedt
- Martini-Klinik, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Anja Wittig
- Proteomedix AG, Zurich-Schlieren, Switzerland
| | - Ramy Huber
- Proteomedix AG, Zurich-Schlieren, Switzerland
| | | | - Ralph Schiess
- Proteomedix AG, Zurich-Schlieren, Switzerland
- * E-mail:
| | - Thomas Steuber
- Martini-Klinik, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Quan Y, Zhang X, Butler W, Du Z, Wang M, Liu Y, Ping H. The role of N-cadherin/c-Jun/NDRG1 axis in the progression of prostate cancer. Int J Biol Sci 2021; 17:3288-3304. [PMID: 34512147 PMCID: PMC8416735 DOI: 10.7150/ijbs.63300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/06/2021] [Indexed: 02/03/2023] Open
Abstract
The dysregulation of androgen receptor (AR) signaling is a critical event in the progression of prostate cancer (PCa) and hormone therapy consisting of androgen deprivation (ADT) or AR inhibition is therefore used to treat advanced cases. It is known that N-cadherin becomes upregulated following ADT and can directly induce PCa transformation to the castration-resistant stage (CRPC). However, the relationship between AR and N-cadherin is unclear and may promote better understanding of CRPC pathogenesis and progression. Here, we demonstrate a new axis of N-cadherin/c-Jun/N-myc downstream regulated gene 1 (NDRG1) that N-cadherin promotes c-Jun expression and suppresses NDRG1 to promote invasion and migration of PCa cells through epithelial to mesenchymal transition (EMT). Targeting N-cadherin in combination with enzalutamide (ENZ) treatment synergistically suppressed PC3 cell proliferation in vivo and in vitro. Further studies showed that compared to lower Gleason score (GS) (GS < 7) cases, high GS (GS > 7) cases exhibited elevated N-cadherin expression and reduced NDRG1 expression, corroborating our in vitro observations. We further demonstrate that c-Jun, AR, and DNA methyltransferase-1 (DNMT1) form a complex in the 12-O-tetradecanoyl phorbol-13-acetate (TPA) response elements (TREs) region of the NDRG1 promoter, which suppresses NDRG1 transcription through DNA hypermethylation. In conclusion, we demonstrate an underlying mechanism for how N-cadherin collaborates with AR and NDRG1 to promote CRPC progression. Controlling N-cadherin/c-Jun/NDRG1 axis may help to overcome resistance to commonly used hormone therapy to improve long-term patient outcomes.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - William Butler
- Department of Pathology, Duke University School of Medicine, Durham NC 27710, USA
| | - Zhen Du
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yuexin Liu
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
12
|
Role of prostate health index to predict Gleason score upgrading and high-risk prostate cancer in radical prostatectomy specimens. Sci Rep 2021; 11:17447. [PMID: 34465825 PMCID: PMC8408259 DOI: 10.1038/s41598-021-96993-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/18/2021] [Indexed: 11/25/2022] Open
Abstract
We evaluated the role of prostate health index (PHI) in predicting Gleason score (GS) upgrading in International Society of Urological Pathology Grade Group (ISUP GG) 1 & 2 prostate cancer (PCa) or adverse pathologic outcomes at radical prostatectomy (RP). A total of 300 patients with prostate specific antigen ≥ 3 ng/mL, PHI and prostate biopsy (71 patients with RP included) were retrospectively included in the study. The primary study outcomes are PCa and clinically significant PCa (csPCa, defined as ISUP GG ≥ 2) diagnostic rate of PHI, and GS upgrading rate at RP specimen. The secondary outcomes are the comparison between GS upgrading and non-upgrading group, GS upgrading and high-risk PCa (ISUP GG ≥ 3 or ≥ pT3a) predictability of preoperative clinical factors. Overall, 139 (46.3%) and 92 (30.7%) were diagnosed with PCa and csPCa, respectively. GS upgrading rate was 34.3% in all patients with RP. Significant differences were shown in the total prostate volume (p = 0.047), the distribution of ISUP GG at biopsy (p = 0.001) and RP (p = 0.032), respectively. PHI values ≥ 55 [Odds ratio (OR): 3.64 (95% confidence interval (CI) = 1.05–12.68, p = 0.042] and presence of PI-RADS lesion ≥ 4 (OR: 7.03, 95% CI = 1.68–29.51, p = 0.018) were the significant predictors of GS upgrading in RP specimens (AUC = 0.737). PHI values ≥ 55 (OR: 9.05, 5% CI = 1.04–78.52, p = 0.046) is a significant factor for predicting adverse pathologic features in RP specimens (AUC = 0.781). PHI could predict GS upgrading in combination with PIRADS lesions ≥ 4 in ISUP GG 1 & 2. PHI alone could evaluate the possibility of high-risk PCa after surgery as well.
Collapse
|
13
|
Rajwa P, Schuettfort VM, D'Andrea D, Quhal F, Mori K, Katayama S, Laukhtina E, Pradere B, Motlagh RS, Mostafaei H, Grossmann NC, Huebner N, Aulitzky A, Mun DH, Briganti A, Karakiewicz PI, Fajkovic H, Shariat SF. Impact of systemic Immune-inflammation Index on oncologic outcomes in patients treated with radical prostatectomy for clinically nonmetastatic prostate cancer. Urol Oncol 2021; 39:785.e19-785.e27. [PMID: 34116934 DOI: 10.1016/j.urolonc.2021.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/18/2021] [Accepted: 05/01/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE To evaluate the predictive and prognostic value of the Systemic Immune-inflammation Index (SII) in a large cohort of patients treated with radical prostatectomy (RP) for clinically non-metastatic prostate cancer (PCa). METHODS We retrospectively analyzed our multicenter database comprising 6,039 consecutive patients. The optimal preoperative SII cut-off value was assessed with the Youden index calculated on a time-dependent receiver operating characteristic (ROC) curve. Logistic regression and Cox regression analyses were used to investigate the association of SII with pathologic features and biochemical recurrence (BCR), respectively. The discriminatory ability of the models was evaluated by calculating the concordance-indices (C-Index). The clinical benefit of the implementation of SII in clinical decision making was assessed using decision curve analysis (DCA). RESULTS Patients with high preoperative SII (≥ 620) were more likely to have adverse clinicopathologic features. On multivariable logistic regression analysis, high preoperative SII was independently associated with extracapsular extension (odds ratio [OR] 1.16, P = 0.041), non-organ confined disease (OR 1.18, P = 0.022), and upgrading at RP (OR 1.23, P < 0.001). We built two Cox regression models including preoperative and postoperative variables. In the preoperative multivariable model, high preoperative SII was associated with BCR (hazard ratio [HR] 1.34, 95% CI 1.15-1.55, P < 0.001). In the postoperative multivariable model, SII was not associated with BCR (P = 0.078). The addition of SII to established models did not improve their discriminatory ability nor did it increase the clinical net benefit on DCA. CONCLUSION In men treated with RP for clinically nonmetastatic PCa, high preoperative SII was statistically associated with an increased risk of adverse pathologic features at RP as well as BCR. However, it did not improve the predictive accuracy and clinical value beyond that obtained by current predictive and prognostic models. SII together with a panel of complementary biomarkers is praised to help guide decision-making in clinically nonmetastatic PCa.
Collapse
Affiliation(s)
- Pawel Rajwa
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Victor M Schuettfort
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David D'Andrea
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Fahad Quhal
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Keiichiro Mori
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Satoshi Katayama
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ekaterina Laukhtina
- Department of Urology, Medical University of Vienna, Vienna, Austria; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Benjamin Pradere
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Reza Sari Motlagh
- Department of Urology, Medical University of Vienna, Vienna, Austria; Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Mostafaei
- Department of Urology, Medical University of Vienna, Vienna, Austria; Research Center for Evidence Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nico C Grossmann
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Nicolai Huebner
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Andreas Aulitzky
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Dong-Ho Mun
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Alberto Briganti
- Department of Urology and Division of Experimental Oncology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, University of Montreal Health Centre, Montreal, Canada
| | - Harun Fajkovic
- Department of Urology, Medical University of Vienna, Vienna, Austria; Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
| | - Shahrokh F Shariat
- Department of Urology, Medical University of Vienna, Vienna, Austria; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia; Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria; Department of Urology, Weill Cornell Medical College, New York, NY USA; Department of Urology, University of Texas Southwestern, Dallas, TX USA; Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
14
|
Eggener S, Karsh LI, Richardson T, Shindel AW, Lu R, Rosenberg S, Goldfischer E, Korman H, Bennett J, Newmark J, Denes BS. A 17-gene Panel for Prediction of Adverse Prostate Cancer Pathologic Features: Prospective Clinical Validation and Utility. Urology 2019; 126:76-82. [DOI: 10.1016/j.urology.2018.11.050] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/06/2018] [Accepted: 11/11/2018] [Indexed: 11/26/2022]
|
15
|
Kaye DR, Qi J, Morgan TM, Linsell S, Ginsburg KB, Lane BR, Montie JE, Cher ML, Miller DC. Pathological upgrading at radical prostatectomy for patients with Grade Group 1 prostate cancer: implications of confirmatory testing for patients considering active surveillance. BJU Int 2018; 123:846-853. [PMID: 30248225 DOI: 10.1111/bju.14554] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine the association between National Comprehensive Cancer Network (NCCN) risk, number of positive biopsy cores, age, and early confirmatory test results on pathological upgrading at radical prostatectomy (RP), in order to better understand whether early confirmatory testing and better risk stratification are necessary for all men with Grade Group (GG) 1 cancers who are considering active surveillance (AS). PATIENTS AND METHODS We identified men in Michigan initially diagnosed with GG1 prostate cancer, from January 2012 to November 2017, who had a RP within 1 year of diagnosis. Our endpoints were: (i) ≥GG2 cancer at RP and (ii) adverse pathology (≥GG3 and/or ≥pT3a). We compared upgrading according to NCCN risk, number of positive biopsy cores, and age. Last, we examined if confirmatory test results were associated with upgrading or adverse pathology at RP. RESULTS Amongst 1966 patients with GG1 cancer at diagnosis, the rates of upgrading to ≥GG2 and adverse pathology were 40% and 59% (P < 0.001), and 10% and 17% (P = 0.003) for patients with very-low- and low-risk cancers, respectively. Upgrading by volume ranged from 49% to 67% for ≥GG2, and 16% to 23% for adverse pathology. Generally, more patients aged ≥70 vs <70 years had adverse pathology. Unreassuring confirmatory test results had a higher likelihood of adverse pathology than reassuring tests (35% vs 18%, P = 0.017). CONCLUSIONS Upgrading and adverse pathology are common amongst patients initially diagnosed with GG1 prostate cancer. Early use of confirmatory testing may facilitate the identification of patients with more aggressive disease ensuring improved risk classification and safer selection of patients for AS.
Collapse
Affiliation(s)
- Deborah R Kaye
- Dow Division of Health Services Research, University of Michigan, Ann Arbor, MI, USA.,University of Michigan, Ann Arbor, MI, USA
| | - Ji Qi
- Dow Division of Health Services Research, University of Michigan, Ann Arbor, MI, USA
| | | | - Susan Linsell
- Dow Division of Health Services Research, University of Michigan, Ann Arbor, MI, USA
| | | | | | - James E Montie
- Dow Division of Health Services Research, University of Michigan, Ann Arbor, MI, USA.,University of Michigan, Ann Arbor, MI, USA
| | - Michael L Cher
- Department of Urology, Wayne State University, Detroit, MI, USA
| | - David C Miller
- Dow Division of Health Services Research, University of Michigan, Ann Arbor, MI, USA.,University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
16
|
A 17-Gene Genomic Prostate Score Assay Provides Independent Information on Adverse Pathology in the Setting of Combined Multiparametric Magnetic Resonance Imaging Fusion Targeted and Systematic Prostate Biopsy. J Urol 2018. [DOI: 10.1016/j.juro.2018.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|