1
|
Gessain G, Lacroix-Triki M. Computational pathology for breast cancer: Where do we stand for prognostic applications? Breast 2025; 81:104464. [PMID: 40179582 PMCID: PMC11999363 DOI: 10.1016/j.breast.2025.104464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/24/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
The very early days of artificial intelligence (AI) in healthcare are behind us. AI is now spreading in the healthcare sector and is gradually being implemented in routine clinical practice. Driven by the increasing digitization of microscope slides, computational pathology (CPath) is further strengthening the role of AI in the field of oncology. CPath is transforming fundamental research as well as routine clinical practice, both for diagnostic and prognostic applications. In breast cancer, CPath holds the potential to address several unmet clinical needs, particularly in the areas of biomarkers and prognostic tools. Indeed, multiple applications are on their way, ranging from predicting clinically meaningful endpoints to offering alternatives to gene-expression testing and detecting molecular alterations directly from digitized whole slide images. However, to fully harness the potential of CPath, several challenges must be overcome. These include improving the availability of multimodal patient data, advancing the digitalization of pathology laboratories, increasing adoption within the medical community, and navigating regulatory hurdles. This review offers an overview of the current landscape of CPath in breast cancer, highlighting the progress made and the hurdles that remain for its widespread clinical adoption in prognostic applications.
Collapse
Affiliation(s)
- Grégoire Gessain
- Department of Pathology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, cedex, 94805, Villejuif, France; Université Paris-Cité, Faculté de Santé, Paris, France
| | - Magali Lacroix-Triki
- Department of Pathology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, cedex, 94805, Villejuif, France.
| |
Collapse
|
2
|
Kishan AU, Lamb JM, Wilhalme H, Casado M, Chong N, Zello L, Juarez JE, Jiang T, Neilsen BK, Low DA, Yang Y, Neylon J, Basehart V, Martin Ma T, Valle LF, Cao M, Steinberg ML. Magnetic Resonance Imaging Versus Computed Tomography Guidance for Stereotactic Body Radiotherapy in Prostate Cancer: 2-year Outcomes from the MIRAGE Randomized Clinical Trial. Eur Urol 2025; 87:622-625. [PMID: 39537438 DOI: 10.1016/j.eururo.2024.10.026] [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: 08/25/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
It has been shown that magnetic resonance imaging (MRI) guidance versus computed tomography (CT) guidance for aggressive margin-reduction (AMR) for stereotactic body radiotherapy (SBRT) in prostate cancer reduces acute toxicity, but the longer-term benefits are unknown. We performed a secondary analysis of MIRAGE, a phase 3 randomized clinical trial of MRI-guided SBRT for prostate cancer, to determine whether AMR with MRI guidance significantly reduced 2-yr physician-scored or patient-reported toxic effects in comparison to CT guidance. The cumulative incidence of 2-yr physician-scored toxicity, defined as grade ≥2 genitourinary (GU) and gastrointestinal (GI) toxic effects according to Common Terminology Criteria for Adverse Events v4.03, were lower with MRI guidance. Cumulative incidence rates of late grade ≥2 toxicity at 2 yr with MRI-guided versus CT-guided SBRT were 27% (95% confidence interval [CI] 19-39%)] versus 51% (95% CI 41-63%) for GU toxicity (p = 0.004), and 1.4% (95% CI 0.2-9.6) versus 9.5% (95% CI 4.6-19) for GI toxicity (p = 0.025). Cumulative logistic regression revealed that MRI-guided SBRT was associated with significantly lower odds of a clinically relevant deterioration in bowel function according to the Expanded Prostate Cancer Index Composite-26 score (odds ratio 0.444, 95% CI 0.209-0.942; p = 0.035) and in the Sexual Health Inventory in Men score (odds ratio 0.366, 95% CI 0.148-0.906; p = 0.03). There were no significant differences in the odds of a deterioration for other quality-of-life metrics. These findings support the hypothesis that aggressive planning for margin reduction for prostate SBRT using MRI leads to continued reductions in toxic effects over 2-yr follow-up. This trial is registered on ClinicalTrials.gov Identifier as NCT04384770.
Collapse
Affiliation(s)
- Amar U Kishan
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA.
| | - James M Lamb
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Holly Wilhalme
- Statistics Core, Department of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Maria Casado
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Natalie Chong
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Lily Zello
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Jesus E Juarez
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Tommy Jiang
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Beth K Neilsen
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Daniel A Low
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Yingli Yang
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - John Neylon
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Vincent Basehart
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Ting Martin Ma
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Luca F Valle
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Minsong Cao
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Michael L Steinberg
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Abida W, Beltran H, Raychaudhuri R. State of the Art: Personalizing Treatment for Patients With Metastatic Castration-Resistant Prostate Cancer. Am Soc Clin Oncol Educ Book 2025; 45:e473636. [PMID: 40112242 DOI: 10.1200/edbk-25-473636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Until recently, the treatment of metastatic castration-resistant prostate cancer (mCRPC) relied exclusively on hormonal therapies and taxane chemotherapy. The advent of modern molecular profiling methods applied in the clinic, namely, next-generation sequencing and advanced positron emission tomography (PET) imaging, has allowed for the development of biomarker-driven therapeutics including anti-PD-L1 therapy for microsatellite instability-high or tumor mutation burden-high disease, poly(ADP-ribose) polymerase (PARP) inhibitors for patients with DNA damage repair mutations, and lutetium 177 vipivotide tetraxetan (177Lu-PSMA-617) for patients with prostate-specific membrane antigen (PSMA) PET-avid disease. While these targeted therapies have improved outcomes, there is an opportunity to refine biomarkers to optimize patient selection, understand resistance, and develop novel combination strategies. In addition, studies in the laboratory and in patient-derived samples have shown that a subset of mCRPC tumors lose expression of common prostate cancer markers such as prostate-specific antigen and PSMA because of lineage plasticity and the development of non-androgen receptor (AR)-driven disease. Non-AR-driven prostate cancer has been associated with aggressive behavior and poor prognosis, including in some cases histologic transformation to a poorly differentiated neuroendocrine prostate cancer (NEPC). The clinical management of NEPC typically follows the treatment paradigm for small cell lung cancer and increasingly relies on genomic and phenotypic characterization of disease, including loss of tumor suppressors and expression of cell surface markers such as DLL3. Therefore, both genomic subtyping and phenotypic subtyping are important to consider and can guide the clinical management of patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruben Raychaudhuri
- University of Washington and the Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
4
|
Losee M, Kavanaugh M, Liu M, Borges N, Haberman V, Ritzer J, Wolanski A, Bhimaniya S, Choudhury AD, Hyun H, Stoltenberg H, Kilbridge KL, Morgans A, Pomerantz M, Robertson M, Sakellis C, Shah H, Taplin ME, Wei XX, Ng T, Ravi P, Jacene H. Outcome of Subsequent Therapies After 177Lu-Vipivotide Tetraxetan for Metastatic Castrate-Resistant Prostate Cancer: A Tertiary Cancer Center Experience. Prostate 2025; 85:742-748. [PMID: 40103237 DOI: 10.1002/pros.24880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND 177Lu-vipivotide tetraxetan (177Lu-PSMA-617, LuPSMA) improves overall survival in patients with metastatic castration-resistant prostate cancer (mCRPC) after at least one taxane chemotherapy and androgen receptor pathway inhibitor. There are limited data on the clinical course and outcomes of patients with mCRPC after receipt of LuPSMA. METHODS We queried an IRB-approved prospectively maintained registry of all patients with mCRPC who received standard-of-care LuPSMA at our institution between June 2022 and January 2024. Clinical data about LuPSMA and subsequent therapies were extracted from the electronic medical record, including the type and number of subsequent systemic therapies, reason for treatment cessation, hematologic toxicity and supportive treatment, and PSA50 response to subsequent therapy (defined as a ≥ 50% decrease in PSA). RESULTS A total of 146 patients were evaluated; mean age 72 (range 52-87), observed median follow-up 5.9 months (range 0.51-18.7). Forty-four received systemic treatment after LuPSMA. The most common subsequent treatment after LuPSMA was chemotherapy (n = 27), primarily cabazitaxel ± carboplatin/cisplatin (n = 23), and the median number of cycles received was 4 (range 1-7). In 35/44 men with available hematologic toxicity data, 13 developed grade ≥ 3 anemia, 7 had ≥ grade 3 thrombocytopenia, and 16 received hematologic support. PSA50 to post-LuPSMA treatment occurred in 10/36 (28%) evaluable patients. Median overall survival from subsequent systemic therapy was 7.6 months (95% CI 5.81-NR). CONCLUSIONS 30% of patients receiving standard-of-care LuPSMA received subsequent therapy, mostly cabazitaxel-containing regimens. Post-LuPSMA treatment appeared tolerable and was associated with a PSA50 response rate of 28%. These outcomes may be biased by limited standard-of-care life-prolonging treatment options at the time of LuPSMA FDA approval, but it also highlights the continued need to develop novel therapeutic strategies for mCRPC post-LuPSMA.
Collapse
Affiliation(s)
- Meryam Losee
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Kavanaugh
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Mofei Liu
- Division of Biostatistics, Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nuno Borges
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Veronica Haberman
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jolivette Ritzer
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Andrew Wolanski
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sudhir Bhimaniya
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Atish D Choudhury
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hyewon Hyun
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Hailey Stoltenberg
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kerry L Kilbridge
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alicia Morgans
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark Pomerantz
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Matthew Robertson
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Christopher Sakellis
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Hina Shah
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Mary-Ellen Taplin
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiao X Wei
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Thomas Ng
- Division of Biostatistics, Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Praful Ravi
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Heather Jacene
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham & Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Carmeli Y, Shpatz Y, Oren-Ivry I, Mansano A, Lewin R, BarOrian I, Mattout J, Weiss I, Haisraely O, Lawrence YR, Symon Z. Effect of Continuous Nutritionist-led Guidance on Bowel Preparation in Patients Undergoing Prostate Stereotactic Body Radiation Treatment With Endorectal Spacing: A Prospective Pilot Trial. Am J Clin Oncol 2025; 48:314-318. [PMID: 39964031 DOI: 10.1097/coc.0000000000001178] [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: 05/23/2025]
Abstract
OBJECTIVE To evaluate the effect of a daily nutritionist consultation on rectal volume, gas, and prostate displacement during Stereotactic Body Radiation Treatment (SBRT) with an endorectal spacer. METHODS Twenty-six consecutive patients receiving 5 fraction SBRT with endorectal spacing were prospectively enrolled for an intensive daily nutritionist intervention utilizing biofeedback based on image guidance from each fraction. A retrospective control cohort receiving a standard bowel preparation was compared. Rectal volume, rectal gas, and prostate displacement were assessed by analysis of cone beam computed tomography. Data was analyzed using the SPSS statistics software. RESULTS Intense dietary intervention with biofeedback led to a consistently lower rectal gas score over 5 fractions ( P <0.001) and less variability in rectal volume during prostate SBRT indicating a nonsignificant trend for superior preparation in the intervention group compared with controls, particularly for the first 2 fractions. However, there was no significant impact on prostate displacement as measured by couch correction. CONCLUSIONS Intense dietary consultations effectively reduce rectal gas and variation of rectal volume during prostate SBRT with endorectal spacing. However, there was no advantage in reducing prostate displacement. Thus, labor-intensive daily nutritionist intervention with biofeedback is not cost-effective in reducing organ motion in patients with endorectal spacers compared with standard pretreatment dietary advice and is not recommended.
Collapse
Affiliation(s)
- Yonatan Carmeli
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University
| | - Yael Shpatz
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Iris Oren-Ivry
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Anat Mansano
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Ron Lewin
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Idan BarOrian
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Jacob Mattout
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Ilana Weiss
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Ory Haisraely
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Yaacov Richard Lawrence
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Zvi Symon
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University
- Department of Radiation Oncology, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
6
|
Wang J, Li L, Feng T, Wang R, Xiang J, Luo Y, Zhu L, Kung HF, Yan W, Zhu Z. Bisphosphonate Bone Imaging Agent [68Ga]Ga-P15-041 for Evaluating Skeletal Metastases in Prostate Cancer: A Comparison With [68Ga]Ga-PSMA-11 PET/CT. Clin Nucl Med 2025; 50:517-522. [PMID: 40320629 DOI: 10.1097/rlu.0000000000005883] [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: 12/18/2024] [Accepted: 02/28/2025] [Indexed: 06/04/2025]
Abstract
BACKGROUND Accurate diagnosis of bone metastases in prostate cancer is essential for staging, prognosis, and treatment. Although PSMA PET/CT is highly effective, complementary imaging is needed to clarify indeterminate lesions. The novel bisphosphonate-based agent [68Ga]Ga-P15-041 shows superior diagnostic accuracy over conventional SPECT imaging, indicating its potential as an auxiliary diagnostic tool. This study explores its role in detecting and assessing prostate cancer bone metastases. PATIENTS AND METHODS This prospective study enrolled 35 patients with prostate cancer and skeletal metastases, who underwent both [68Ga]Ga-P15-041 and [68Ga]Ga-PSMA-11 PET/CT within 1 week. Lesions detected by [68Ga]Ga-PSMA-11 PET/CT were classified using Prostate-specific Membrane Antigen Reporting and Data System 2.0. RESULTS [68Ga]Ga-P15-041 PET/CT detected more lesions than [68Ga]Ga-PSMA-11 PET/CT (525 vs 509, P < 0.001) and demonstrated significantly higher tracer uptake, with a mean SUV of 20.73 ± 14.67 compared with 11.13 ± 8.12 (P < 0.0001). It detected significantly more osteoblastic lesions (504 vs 391, P < 0.0001). In addition, this study established the Reporting and Data System for [68Ga]Ga-P15-041 (P15-041-RADS), which classifies prostate cancer bone metastases into 5 categories based on SUVmax and morphologic changes. P15-041-RADS reclassified 85.71% of Prostate-specific Membrane Antigen Reporting and Data System category 3 lesions and 95.00% of 5T lesions into higher-confidence categories, offering improved diagnostic clarity. Limitations include small sample size and lack of pathologic gold standards. CONCLUSIONS [68Ga]Ga-P15-041 PET/CT is a promising and accessible bone imaging agent that could complement [68Ga]Ga-PSMA-11 PET/CT in the diagnosis and classification of bone metastases in prostate cancer.
Collapse
Affiliation(s)
- Jiarou Wang
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College
- State Key Laboratory of Complex Severe and Rare Diseases
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine
| | - Linlin Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College
- State Key Laboratory of Complex Severe and Rare Diseases
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine
| | - Tianrui Feng
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College
| | - Rongxi Wang
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College
- State Key Laboratory of Complex Severe and Rare Diseases
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine
| | - Jialin Xiang
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College
- State Key Laboratory of Complex Severe and Rare Diseases
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine
| | - Yaping Luo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College
- State Key Laboratory of Complex Severe and Rare Diseases
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine
| | - Lin Zhu
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Ministry of Education, Beijing, China
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Weigang Yan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College
- State Key Laboratory of Complex Severe and Rare Diseases
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine
| |
Collapse
|
7
|
Xu J, Lu J, Gielzak M, Lilly Zheng S, Lu L, Wei J, Cornell B, Shi Z, Wang Q, Tran H, Engelmann V, Ashworth A, Lin K, Ross AE, Walsh PC, Marshall C, Luo J, Isaacs WB, Helfand BT, Pavlovich CP. Germline Testing for Prostate Cancer Patients: Evidence-Based Evaluation of Genes Recommended by NCCN Guidelines. Prostate 2025. [PMID: 40405569 DOI: 10.1002/pros.24918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Approximately 50% of prostate cancer (PCa) patients meet the National Comprehensive Cancer Network (NCCN) guidelines for germline testing at diagnosis. However, the selection of genes for testing, their supporting evidence, and clinical interpretation remain poorly understood. METHODS An evidence-based evaluation of the recommended genes was conducted using data from the UK Biobank and Johns Hopkins School of Medicine, including 22,052 PCa patients and 191,055 unaffected controls. Association of germline pathogenic/likely pathogenic (P/LP) variants in each gene was tested using logistic regression, adjusting for age and genetic background. RESULTS Among the 11 NCCN-recommended PCa-related genes, significant associations (p < 0.0045) were identified between germline P/LP variants of five genes (HOXB13, BRCA2, ATM, CHEK2, and MSH2) and PCa risk. Additionally, BRCA2 and ATM variants were significantly associated with PCa aggressiveness. Of the 19 NCCN-recommended genes related to PARPi sensitivity, consistent evidence supported an enhanced response to PARPi therapy in patients with BRCA2 alterations, with weaker evidence for BRCA1, and limited supporting evidence for the remaining genes. Germline P/LP variants in BRCA2 and BRCA1 were observed in 0.77% and 0.14% of unselected PCa patients, respectively. Notably, no published study specifically assessed the efficacy of germline alterations, which were considerably rarer than somatic mutations. CONCLUSION Supporting statistical evidence is available for only a subset of the NCCN-recommended genes for germline testing. This evidence-based analysis may aid urologists-particularly those without specialized genetics training-in understanding germline testing for PCa risk assessment, prognosis, and treatment decision-making in clinical practice.
Collapse
Affiliation(s)
- Jianfeng Xu
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
- Division of Urology, Department of Surgery, Endeavor Health, Evanston, Illinois, USA
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | - Jim Lu
- GoPath Lab, LLC, Buffalo Grove, Illinois, USA
| | - Marta Gielzak
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | - S Lilly Zheng
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | - Lucy Lu
- GoPath Lab, LLC, Buffalo Grove, Illinois, USA
| | - Jun Wei
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | | | - Zhuqing Shi
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | - Qiang Wang
- GoPath Lab, LLC, Buffalo Grove, Illinois, USA
| | - Huy Tran
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | | | - Annabelle Ashworth
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | - Kirk Lin
- Arizona Urology Specialists, Phoenix, Arizona, USA
| | - Ashley E Ross
- Department of Urology, Northwestern University Feinberg School of Medicine Northwestern, Chicago, Illinois, USA
| | - Patrick C Walsh
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | | | - Jun Luo
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | - William B Isaacs
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | - Brian T Helfand
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
- Division of Urology, Department of Surgery, Endeavor Health, Evanston, Illinois, USA
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | - Christian P Pavlovich
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| |
Collapse
|
8
|
Cooperberg MR. Prostate Cancer Active Surveillance Without Biopsy? Not Quite Yet, but Watch This Space. Eur Urol 2025:S0302-2838(25)00286-6. [PMID: 40393863 DOI: 10.1016/j.eururo.2025.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/22/2025]
Affiliation(s)
- Matthew R Cooperberg
- Departments of Urology and Epidemiology & Biostatistics, University of California-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
9
|
Lee J, Kim T. Current Status and Future Perspectives of Nuclear Medicine in Prostate Cancer from Imaging to Therapy: A Comprehensive Review. Biomedicines 2025; 13:1132. [PMID: 40426959 PMCID: PMC12109171 DOI: 10.3390/biomedicines13051132] [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: 03/28/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Nuclear medicine has emerged as a critical modality in the diagnostic and therapeutic management of urological malignancies, particularly prostate cancer. Advances in single-photon emission computed tomography/computed tomography (CT) and positron emission tomography/CT (PET/CT) have enhanced tumor assessment across staging, treatment response, and recurrence settings. Molecular imaging, which offers insights beyond traditional anatomical imaging, is increasingly integral in specific clinical scenarios. Theranostic nuclear medicine, which combines diagnostic imaging with targeted therapy, has become a well-established treatment option, particularly for patients with metastatic castration-resistant prostate cancer (mCRPC). The development of the prostate-specific membrane antigen (PSMA) radioligands has revolutionized clinical management by enabling precise disease staging and delivering effective radioligand therapy (RLT). Ongoing research aims to refine the role of PSMA PET imaging in staging and treatment monitoring, while optimizing PSMA-targeted RLT for broader clinical use. Given that prostate cancer remains highly prevalent, the anticipated increase in the demand for RLT presents both challenges and opportunities for nuclear medicine services globally. Theranostic approaches exemplify personalized medicine by enabling the tailoring of treatments to individual tumor biology, thereby improving survival outcomes and maintaining patients' quality of life with minimal toxicity. Although the current focus is on advanced disease, future research holds promise for expanding these strategies to earlier stages, potentially enhancing curative prospects. This evolving field not only signifies a paradigm shift in the care of prostate cancer patients but also underscores the growing importance of nuclear medicine in delivering precision oncology.
Collapse
Affiliation(s)
- Joohee Lee
- CHA Ilsan Medical Center, Department of Nuclear Medicine, CHA University College of Medicine, Ilsan 10414, Gyeonggi-do, Republic of Korea;
| | - Taejin Kim
- CHA Ilsan Medical Center, Department of Urology, CHA University College of Medicine, Ilsan 10414, Gyeonggi-do, Republic of Korea
| |
Collapse
|
10
|
Mandl A, Zahurak ML, Metri NA, Shore ND, Mao S, McKay RR, Taplin ME, Szmulewitz RZ, Maughan BL, Reichert ZR, Kessler ER, Heath EI, Dreicer R, Stein CA, Milne GL, Sfanos KS, Ernst SE, Mummert LA, Cruz-Lebrón A, Michel SLJ, Kane MA, Hursey M, Worth MA, Wagner WD, Eshleman JR, Debeljak M, Xu L, Cao H, Dowling D, Marshall CH, Markowski MC, Denmeade SR, Eisenberger MA, Antonarakis ES, Carducci MA, Paller CJ. Muscadine Grape Skin Extract in Biochemically Recurrent Prostate Cancer: A Randomized, Placebo-Controlled, Biomarker-Enriched Trial in Patients With the SOD2 Ala/Ala Variant. Prostate 2025. [PMID: 40325900 DOI: 10.1002/pros.24903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Many patients with biochemically recurrent prostate cancer (BCRPC) prefer to delay androgen deprivation therapy (ADT) due to its adverse effects, highlighting the need for better-tolerated, effective alternatives. A subgroup analysis of our prior Phase II trial showed that muscadine grape skin extract (MPX) increased PSA doubling time (PSADT) in patients with SOD2 Ala/Ala variant which provided the rationale for this trial. METHODS This randomized, double-blind, placebo-controlled trial, conducted at 14 sites, evaluated patients with BCRPC and SOD2 Ala/Ala genotype. Patients received 4000 mg MPX or placebo daily. The primary endpoint was on-study PSA slope with comparisons between treatment arms. Secondary endpoints were PSADT, PSA response (≥ 50% decrease), and PSA progression free survival (PFS). Correlative studies included markers of oxidative stress and gastrointestinal microbiota composition. RESULTS At interim analysis, fifty-nine patients were randomized (MPX, n = 29; placebo, n = 30). On-study PSA slopes at 12, 24, 36, and 48 weeks showed no significant differences between the MPX and placebo arms (p = 0.49). The study was stopped due to futility. No significant differences were observed in PSADT, PSA response, median PSA PFS, or oxidative stress biomarkers. MPX was well-tolerated, with no grade 3-4 AEs attributable to the study drug. Microbiome analysis showed no significant differences in alpha diversity but revealed increased relative abundance of Roseburia faecis and Akkermansia muciniphila in the MPX group. CONCLUSIONS Although MPX supplementation had no significant effect on PSA slope in men with BCRPC and SOD2 Ala/Ala variant, this study provides a rigorous evaluation of a natural product and highlights the importance of well-designed clinical trials in advancing evidence-based integrative oncology. TRIAL REGISTRATION ClinicalTrials. gov, NCT03535675.
Collapse
Affiliation(s)
- A Mandl
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - M L Zahurak
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - N A Metri
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - N D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - S Mao
- Allegheny Health Network, Pittsburgh, PA, USA
| | - R R McKay
- University of California, San Diego, La Jolla, CA, USA
| | - M E Taplin
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - B L Maughan
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| | - Z R Reichert
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - E R Kessler
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - E I Heath
- Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - R Dreicer
- University of Virginia School of Medicine, Charlottesville, VA, USA
| | - C A Stein
- City of Hope Medical Center, Madras, OR, USA
| | - G L Milne
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - K S Sfanos
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S E Ernst
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - L A Mummert
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - A Cruz-Lebrón
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - M A Kane
- University of Maryland, College Park, MD, USA
| | - M Hursey
- University of Maryland, College Park, MD, USA
| | - M A Worth
- University of Maryland, College Park, MD, USA
| | - W D Wagner
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Muscadine Naturals Inc., Clemmons, NC, USA
| | - J R Eshleman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - M Debeljak
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - L Xu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - H Cao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - D Dowling
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - C H Marshall
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - M C Markowski
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - S R Denmeade
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - M A Eisenberger
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - E S Antonarakis
- University of Minnesota, Masonic Cancer Center, Minneapolis, MN, USA
| | - M A Carducci
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| | - C J Paller
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
11
|
Gnanapragasam VJ. Optimizing clinical risk stratification of localized prostate cancer. Curr Opin Urol 2025:00042307-990000000-00247. [PMID: 40314071 DOI: 10.1097/mou.0000000000001294] [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: 05/03/2025]
Abstract
PURPOSE OF REVIEW To review the current risk and prognostic stratification systems in localised prostate cancer. To explore some of the most promising adjuncts to clinical models and what the evidence has shown regarding their value. RECENT FINDINGS There are many new biomarker-based models seeking to improve, optimise or replace clinical models. There are promising data on the value of MRI, radiomics, genomic classifiers and most recently artificial intelligence tools in refining stratification. Despite the extensive literature however, there remains uncertainty on where in pathways they can provide the most benefit and whether a biomarker is most useful for prognosis or predictive use. Comparisons studies have also often overlooked the fact that clinical models have themselves evolved and the context of the baseline used in biomarker studies that have shown superiority have to be considered. SUMMARY For new biomarkers to be included in stratification models, well designed prospective clinical trials are needed. Until then, there needs to be caution in interpretation of their use for day-to-day decision making. It is critical that users balance any purported incremental value against the performance of the latest clinical classification and multivariate models especially as the latter are cost free and widely available.
Collapse
Affiliation(s)
- Vincent J Gnanapragasam
- Department of Surgery, University of Cambridge
- Cambridge Prostate Cancer and Clinical Trials Group
- Cambridge University Hospitals, Urology, Cambridge, UK
| |
Collapse
|
12
|
Handa N, Bennett R, Li EV, Ho A, Huang MM, Kumar S, Neill C, Alam R, Patel HD, Schaeffer EM, Ross AE. PSMA PET/CT findings in high-risk biochemical recurrence after local treatment of prostate cancer. BJUI COMPASS 2025; 6:BCO270028. [PMID: 40356610 PMCID: PMC12066941 DOI: 10.1002/bco2.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Objectives To describe PSMA PET/CT characteristics of patients with high-risk BCR. Subjects/patients and methods This was a retrospective analysis of patients with high-risk BCR prostate cancer (PSA ≥ 2 ng/ml above nadir after radiation therapy [RT] or ≥1 ng/ml after radical prostatectomy [RP] +/- RT) who underwent PET/CT from July 2021-March 2023. Patients with prior cytotoxic chemotherapy, androgen deprivation therapy (ADT) initiated >3 months prior to PET/CT or positive conventional imaging within 3 months of PET/CT were excluded. Neoadjuvant/adjuvant ADT completed ≥9 months prior was allowed. Logistic regression, Pearson's Chi-squared, Wilcoxon rank sum and Fisher's exact tests were used for analysis. Results A total of 113 of 145 (77%) included patients in the analysis had ≥1 lesion on PSMA PET/CT. There was no difference in PSMA PET/CT positivity based on age, race, Gleason Grade at initial biopsy or PSA. Overall, 29 (20%) patients had lesions in the prostate/prostate bed only, 31 (21%) had lesions consistent with N1M0 disease and 53 (37%) had lesions consistent with M1 disease. For M1 patients, 21/53 (40%) had oligometastatic disease (1-3 lesions), and 32/53 (60%) had a higher burden (>3 lesions). Local recurrence was more common with RT and nodal recurrence with RP, with no difference in distant metastasis by initial treatment. Conclusion Nearly 80% of patients with high-risk BCR after local treatment for prostate cancer with RP and/or RT will have positive findings on PSMA PET/CT. In addition to intensified systemic therapy, up to 55% of the patients may have benefitted from salvage local therapy, nodal pelvic radiation or metastasis-directed therapies for oligometastatic disease.
Collapse
Affiliation(s)
- Nicole Handa
- Northwestern University, Feinberg School of MedicineChicagoILUSA
| | | | - Eric V. Li
- Northwestern University, Feinberg School of MedicineChicagoILUSA
| | - Austin Ho
- Northwestern University, Feinberg School of MedicineChicagoILUSA
| | | | - Sai Kumar
- Northwestern University, Feinberg School of MedicineChicagoILUSA
| | - Clayton Neill
- Northwestern University, Feinberg School of MedicineChicagoILUSA
| | - Ridwan Alam
- Northwestern University, Feinberg School of MedicineChicagoILUSA
| | - Hiten D. Patel
- Northwestern University, Feinberg School of MedicineChicagoILUSA
- Surgery Service, Jesse Brown VA Medical CenterChicagoILUSA
| | | | - Ashley E. Ross
- Northwestern University, Feinberg School of MedicineChicagoILUSA
| |
Collapse
|
13
|
Ghoreifi A, Ramacciotti LS, Kaneko M, Medina LG, Cacciamani GE, Konganige S, Aron M, Sadeghi S, Jadvar H, Djaladat H, Sotelo R, Desai MM, Gill IS, Aron M, Abreu AL. The outcomes of salvage robotic radical prostatectomy following radiation versus focal therapy: Does the primary treatment modality matter? BJUI COMPASS 2025; 6:e70019. [PMID: 40322501 PMCID: PMC12045929 DOI: 10.1002/bco2.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Objectives We aim to compare salvage robotic radical prostatectomy (sRRP) for recurrent prostate cancer (PCa) after primary radiation (RT) versus focal therapy (FT). Materials and Methods Patients who underwent sRRP following primary local therapy for PCa were identified. Perioperative findings and functional/oncologic outcomes were compared in RT versus FT groups. Results Overall, 112 patients were included, with 84 receiving RT and 28 FT as primary treatment. Median age and PSA were 68 years and 5.4 ng/mL, respectively. There was one rectal injury in the RT group. The overall 90-day complications were significantly higher in RT group (33% vs. 11%, p = 0.03). On multivariable analysis, history of RT and prolonged operative time were associated with a higher rate of 90-day complications. The 6- and 12-month continence rates were higher in FT group (50% vs. 20%, p = 0.02 and 69% vs. 33%, p = 0.03). Potency at 12 months was better preserved in FT group (46% vs. 12%, p = 0.01). On final sRRP pathology, the rates of grade group ≥ 4 (51% vs. 36%, p = 0.2), pT3 (69% vs. 75%, p = 0.6), positive nodes (30% vs. 18%, p = 0.2) and positive margins (33% vs. 39%, p = 0.5) were similar for RT versus FT, respectively. The 3-year biochemical recurrence-free survival was 86% for RT versus 94% for FT (p = 0.6).Conclusion: sRRP for recurrent PCa after FT is associated with lower complications and higher urinary continence and potency rates than patients who received primary RT.
Collapse
Affiliation(s)
- Alireza Ghoreifi
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Lorenzo Storino Ramacciotti
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Center for Image‐Guided Surgery, Focal Therapy and Artificial Intelligence for Prostate Cancer, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Masatomo Kaneko
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Center for Image‐Guided Surgery, Focal Therapy and Artificial Intelligence for Prostate Cancer, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Luis G. Medina
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Giovanni E. Cacciamani
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Shiran Konganige
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Manju Aron
- Department of Pathology Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Sarmad Sadeghi
- Department of Medical OncologyUniversity of Southern California Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Hossein Jadvar
- Department of Radiology Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Hooman Djaladat
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Rene Sotelo
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mihir M. Desai
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Inderbir S. Gill
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Center for Image‐Guided Surgery, Focal Therapy and Artificial Intelligence for Prostate Cancer, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Monish Aron
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Andre Luis Abreu
- USC Institute of Urology, Catherine and Joseph Aresty Department of Urology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Center for Image‐Guided Surgery, Focal Therapy and Artificial Intelligence for Prostate Cancer, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
14
|
Turco F, Gillessen S, Herrmann K, Paone G, Omlin A. Treatment Landscape of Prostate Cancer in the Era of PSMA Radiopharmaceutical Therapy. J Nucl Med 2025; 66:665-672. [PMID: 40015917 DOI: 10.2967/jnumed.124.267730] [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: 12/04/2024] [Accepted: 01/29/2025] [Indexed: 03/01/2025] Open
Abstract
The treatment landscape of prostate cancer is quite complex because of the many therapeutic options available in different disease settings (hormonal treatments, chemotherapy, poly(adenosine diphosphate ribose) polymerase inhibitors, radiopharmaceutical therapy). Since in most cases we do not have comparative studies between these different agents, the best therapeutic sequence in patients with prostate cancer remains unsolved. In this review, we describe the different systemic therapeutic options available in each disease setting from localized disease to metastatic castration-resistant disease. We also indicate when to use each of these therapeutic options in the therapeutic sequence on the basis of the results of the available studies. A special focus of this review is the place of prostate-specific membrane antigen radiopharmaceutical therapy in the treatment algorithms.
Collapse
Affiliation(s)
- Fabio Turco
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland;
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium, University Hospital Essen, Essen, Germany
| | - Gaetano Paone
- Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland
- Clinic of Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; and
| | - Aurelius Omlin
- Onkozentrum Zurich, University of Zurich and Tumorzentrum Hirslanden Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
King MT, Merrick GS, Galbreath RW, Fiano R, Butler WM, Wallner KE, Orio PF. Prospective Evaluation of Supplemental External Beam Radiation Therapy With Palladium-103 Prostate Brachytherapy: Long-Term Results of the 44/20/0 Trials. Pract Radiat Oncol 2025; 15:e276-e285. [PMID: 39447863 DOI: 10.1016/j.prro.2024.10.005] [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: 08/21/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
PURPOSE The 44/20 and 20/0 randomized trials evaluated whether different external beam radiation therapy (EBRT) dosing regimens prior to brachytherapy affected biochemical failure (BF). We report long-term outcomes of both trials and evaluate whether biological equivalent dose (BED) was associated with reduced BF in the combined trial cohort. METHODS AND MATERIALS Both trials enrolled patients with clinical T1c to T2b, Gleason scores 7 to 9, and/or a pretreatment prostate-specific antigen (PSA) 10 to 20 ng/mL disease. The 44/20 trial randomized patients to 44 Gy EBRT with 90 Gy palladium (Pd)-103 versus 20 Gy EBRT with 115 Gy Pd-103. The subsequent 20/0 trial randomized patients to the 20 Gy arm versus monotherapeutic 125 Gy Pd-103. For each trial, univariate Fine-Gray analysis evaluated whether the treatment arm was associated with BF for the entire cohort and the unfavorable intermediate-risk (UIR) subgroup. For the combined trial cohort, multivariate Fine-Gray analysis evaluated whether BED was associated with BF while adjusting for clinical factors. RESULTS There were 247 analyzable patients in the 44/20 trial. At a median follow-up of 13.7 years, there were no differences in BF for the entire cohort (subdistribution hazard ratio [sHR] 0.99; 95% CI, 0.43, 2.276; P = .97) or the UIR subgroup (sHR 0.72; 95% CI, 0.25, 2.08; P = .55). There were 383 analyzable patients in the 20/0 trial. At a median follow-up of 10.4 years, there were no differences in BF for the entire cohort (sHR 0.42; 95% CI, 0.13-1.80; P = .15) or the UIR subgroup (sHR 0.81; 95% CI, 0.16-4.03; P = .80). For the combined cohort (630 patients), BED was not associated with BF (1.00; 95% CI, 0.98-1.02; P = .88) on multivariate analyses while adjusting for androgen deprivation therapy utilization, 4-tiered National Comprehensive Cancer Network category, and year of treatment. CONCLUSIONS Brachytherapy monotherapy should be a standard-of-care treatment for clinically localized, intermediate-risk prostate cancer, including UIR disease.
Collapse
Affiliation(s)
- Martin T King
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts.
| | - Gregory S Merrick
- Urologic Research Institute, Sarasota, Florida; Bethany College, Bethany, West Virginia
| | - Robert W Galbreath
- Urologic Research Institute, Sarasota, Florida; Ohio University Eastern, St. Clairsville, Ohio
| | - Ryan Fiano
- Urologic Research Institute, Sarasota, Florida
| | | | - Kent E Wallner
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Peter F Orio
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
16
|
Juweid ME, Al‐Qasem SF, Khuri FR, Gallamini A, Lohmann P, Ziellenbach H, Mottaghy FM. Beyond fluorodeoxyglucose: Molecular imaging of cancer in precision medicine. CA Cancer J Clin 2025; 75:226-242. [PMID: 40183513 PMCID: PMC12061632 DOI: 10.3322/caac.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer molecular imaging is the noninvasive visualization of a process unique to or altered in neoplasia, such as proliferation, glucose metabolism, and receptor expression, which is relevant to patient management. Several molecular imaging modalities are now available, including magnetic resonance, optical, and nuclear imaging. Nuclear imaging, particularly using fluorine-18-fluorodeoxyglucose positron emission tomography, is widely used in the staging and response assessment of multiple cancer types. However, at this writing, new nuclear medicine probes, especially positron emission tomography tracers, are increasingly used or are being investigated for cancer evaluation. This review focuses on these probes, their biologic targets, and the applications or potential applications for their use in the assessment of various neoplasms, including both probes available for commercial use-such as somatostatin receptor ligands in neuroendocrine tumors, prostate-specific membrane antigen ligands in prostate cancer, norepinephrine analogs in neural crest tumors like neuroblastoma, and estrogen analogs in breast cancer-and others in clinical development, such as fibroblast-activating protein inhibitors, C-X-C chemokine receptor type 4 ligands, and monoclonal antibodies targeting receptor tyrosine kinases, CD4-positive or CD8-positive tumor-infiltrating lymphocytes, tumor-associated macrophages, and cancer stem cell biomarkers. These developments represent a major step toward the integration of molecular imaging as a powerful tool in precision medicine, with an expectedly significant impact on patient management and outcome.
Collapse
Affiliation(s)
- Malik E. Juweid
- Department of Radiology and Nuclear MedicineSchool of MedicineUniversity of JordanAmmanJordan
- The National Center for Diabetes, Endocrinology, and GeneticsUniversity of JordanAmmanJordan
| | - Soud F. Al‐Qasem
- Department of Radiology and Nuclear MedicineSchool of MedicineUniversity of JordanAmmanJordan
| | - Fadlo R. Khuri
- Division of OncologyDepartment of Internal MedicineAmerican University of BeirutBeirutLebanon
| | - Andrea Gallamini
- Research and Innovation DepartmentAntoine Lacassagne Cancer CenterNiceFrance
| | - Philipp Lohmann
- Department of Nuclear MedicineUniversity Hospital AachenRWTH Aachen UniversityAachenGermany
- Medical Imaging Physics (INM‐4)Institute of Neuroscience and Medicine, Research Center JuelichJuelichGermany
| | | | - Felix M. Mottaghy
- Department of Nuclear MedicineUniversity Hospital AachenRWTH Aachen UniversityAachenGermany
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtthe Netherlands
- Center of Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)CologneGermany
| |
Collapse
|
17
|
Duque Santana V, Moreno Perez I, Sanmamed Salgado N. Current role of salvage radiotherapy in oligometastatic prostate cancer at the time of recurrence. Curr Opin Urol 2025; 35:308-312. [PMID: 40035188 DOI: 10.1097/mou.0000000000001273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
PURPOSE OF REVIEW To review the evidence for metastasis-directed therapy (MDT) in oligometastatic (OM) prostate cancer (PC) and future directions in this clinical setting. RECENT FINDINGS The indications for radiotherapy in PC have significantly increased over the last decade. The treatment of OM disease has been gaining popularity thanks to new molecular imaging techniques that allow more accurate identification of OM patients who may benefit from MDT. stereotactic body radiation therapy can offer potentially curative treatment with low toxicity rates. Although most published MDT studies focus on hormone-sensitive (HS) PC in the setting of recurrence, promising results have emerged in recent years even in patients with castration resistance. SUMMARY MDT has emerged as an effective treatment option for OM PC patients with favorable oncological outcomes and a low toxicity profile. Evidence suggests that MDT can be used to delay androgen deprivation therapy or in combination with systemic therapy to potentially improve efficacy outcomes. Molecular stratification is needed to determine who will benefit from treatment intensification with MDT +/- systemic therapies.
Collapse
Affiliation(s)
- Victor Duque Santana
- Department of Radiation Oncology, Quironsalud Madrid University Hospital
- Department of Medicine, Faculty of Medicine, Health and Sports, European University of Madrid
| | - Ignacio Moreno Perez
- Department of Medical Oncology, Hospital Clinico Universitario San Carlos
- Investigation Institute, Clinico San Carlos Hospital
| | - Noelia Sanmamed Salgado
- Investigation Institute, Clinico San Carlos Hospital
- Department of Radiation Oncology, Hospital Clinico Universitario San Carlos, Madrid, Spain
| |
Collapse
|
18
|
Rifai S, Rifai A, Shi X, Khan MA, Guang W, Wang L, Tallon L, Hussain A. Genomic and transcriptomic sequencing in prostate cancer. Curr Opin Oncol 2025; 37:240-249. [PMID: 40071471 DOI: 10.1097/cco.0000000000001136] [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: 04/05/2025]
Abstract
PURPOSE OF REVIEW Genomic and transcriptomic sequencing technologies have revolutionized our ability to characterize prostate cancer at the molecular level. The underlying premise of next-generation sequencing technologies and their current and evolving applications in prostate cancer management are provided in the review. RECENT FINDINGS Improved methodologies are allowing timely sequencing of the coding regions or both the coding and noncoding regions of the genome to help identify potential mutations and structural variations in the prostate cancer genome, some of which are currently also targetable therapeutically. DNA microarray- based differential gene expression has been supplanted by RNA sequencing (RNA-seq), which not only allows for more accurate quantitation but also nucleotide-level resolution to investigate the entire transcriptome, including alternative gene spliced transcripts and noncoding RNA transcripts, whose full clinical implications have yet to be fully understood and realized. Gene classifier platforms that predict risk of recurrence or metastasis are being incorporated into prostate cancer management algorithms. In the appropriate clinical context, not only somatic but also germline mutation testing is being recommended. SUMMARY Continued clinical integration of sequencing technologies and ongoing research will lead to improved understanding of prostate cancer biology and prostate cancer treatment.
Collapse
Affiliation(s)
- Safiullah Rifai
- University of Maryland Greenebaum Comprehensive Cancer Center
| | - Azimullah Rifai
- University of Maryland Greenebaum Comprehensive Cancer Center
| | - Xiaolei Shi
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
| | | | - Wei Guang
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
| | - Linbo Wang
- University of Maryland Greenebaum Comprehensive Cancer Center
| | | | - Arif Hussain
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
- Department of Pathology
- Depepartment of Biochemistry and Molecular Biology
- Baltimore VA Medical Center, Baltimore, Maryland USA
| |
Collapse
|
19
|
Monda SM, Demus T, Jaime-Casas S, Meah S, Srivastava A, Sarle R, Labardee C, Ghani KR, Ginsburg KM, Morgan TM, Borza T. Trends in Surgical Overtreatment of Prostate Cancer. JAMA Oncol 2025:2833303. [PMID: 40292607 PMCID: PMC12038712 DOI: 10.1001/jamaoncol.2025.0963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/03/2025] [Indexed: 04/30/2025]
Abstract
Importance Overtreatment of prostate cancer is a public health concern that undermines prostate cancer screening efforts. Objective To assess trends in pathologic grade on prostatectomy during the past 2 decades as a surrogate for overtreatment. Design, Setting, and Participants This retrospective cohort study examined the grade of prostate cancer on final pathology reports among patients undergoing prostatectomy between January 1, 2010, and September 1, 2024, in 2 parallel cohorts: Surveillance, Epidemiology, and End Results (SEER), a nationwide cancer registry, and Michigan Urological Surgery Improvement Collaborative (MUSIC), a statewide clinical registry. The presence of higher-risk features among patients who underwent grade group 1 prostatectomy during this period was also assessed. Exposures The primary exposure of interest was year of radical prostatectomy. Main Outcomes and Measures The primary outcome was the proportion of all prostatectomies that were pathologic grade group 1 (pGG1) on final pathology reports. The secondary outcome was the proportion of pGG1 prostatectomies with a higher-risk preoperative feature, assessed as a binary variable and including at least 1 of the following: more than 50% of biopsy cores positive, prostate-specific antigen of 10 ng/mL or higher, or grade group 2 on biopsy. Results A total of 162 558 male patients in SEER (median [IQR] age, 63 [57-67] years) and 23 370 in MUSIC (median [IQR] age, 64 [59-69] years) underwent prostatectomy. The proportion of radical prostatectomies resulting in pGG1 on final pathology reports decreased from 32.4% (5852 of 18 071) to 7.8% (978 of 12 500) between 2010 and 2020 in SEER and from 20.7% (83 of 401) to 2.7% (32 of 1192) between 2012 and 2024 in MUSIC. A more recent prostatectomy was associated with a lower likelihood of a pGG1 prostatectomy while controlling for age and race within SEER (odds ratio [OR] per 5 years, 0.41; 95% CI, 0.40-0.42; P < .001) and MUSIC (OR per 5 years, 0.39; 95% CI, 0.36-0.43; P < .001). Within a subset analysis of those prostatectomies that were final pGG1, a more recent prostatectomy was associated with the presence of a higher-risk preoperative feature, including more than 50% of biopsy cores positive, prostate-specific antigen of 10 ng/mL or higher, and grade group 2 on prior biopsy within SEER (OR per 5 years, 1.60; 95% CI, 1.54-1.67; P < .001) and MUSIC (OR per 5 years, 1.60; 95% CI, 1.34-1.90; P < .001). Conclusions and Relevance This cohort study found that since 2010, the frequency of pGG1 prostatectomies markedly decreased, and those few that were performed were more likely to have a higher-risk feature. This reduction in the proportion of prostatectomies that are pGG1 likely reflects improved diagnostic pathways, adherence to active surveillance protocols for low-risk cases, and ongoing efforts at both the state and national levels to minimize unnecessary surgical interventions in patients diagnosed with clinically insignificant prostate cancer.
Collapse
Affiliation(s)
| | - Timothy Demus
- Department of Urology, Sparrow Health System, Lansing, Michigan
| | - Salvador Jaime-Casas
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Sabir Meah
- Department of Urology, University of Michigan, Ann Arbor
| | | | - Richard Sarle
- Department of Urology, Sparrow Health System, Lansing, Michigan
| | | | | | | | - Todd M. Morgan
- Department of Urology, University of Michigan, Ann Arbor
| | - Tudor Borza
- Department of Urology, University of Michigan, Ann Arbor
| |
Collapse
|
20
|
Huang S, Yin H. Exploring the Molecular Mechanism and Role of Glutathione S-Transferase P in Prostate Cancer. Biomedicines 2025; 13:1051. [PMID: 40426879 PMCID: PMC12109251 DOI: 10.3390/biomedicines13051051] [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: 03/01/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Aims: To investigate the effect of Glutathione metabolism in prostate cancer pathogenesis. Background: There is growing evidence that Glutathione metabolism plays an important role in prostate cancer, with genes encoding key enzymes in this pathway potentially serving as diagnostic or prognostic biomarkers. Objective: To explore whether there is a causal relationship between key enzymes in the Glutathione metabolism and prostate cancer, and to further investigate the molecular mechanisms and roles of the genes encoding their proteins in relation to prostate cancer. Method: Transcriptomic datasets from the Gene Expression Omnibus (GEO) database were analyzed to identify differentially expressed genes (DEGs) and enriched pathways in prostate cancer versus normal tissues. Two-sample bidirectional Mendelian randomization (MR) was employed to assess causal relationships between Glutathione metabolic enzymes (exposure) and prostate cancer risk (outcome). Immune infiltration analysis and LASSO regression were performed to construct a diagnostic model. Single-cell RNA sequencing (scRNA-seq) data were utilized to elucidate cell-type-specific expression patterns and functional associations of target genes. Result: The results of two-sample bidirectional MR showed that Glutathione S-transferase P (GSTP) in Glutathione metabolism could reduce the risk of prostate cancer. The Glutathione S-transferase Pi-1 (GSTP1) gene was lowly expressed in prostate cancer and was able to diagnose prostate cancer more accurately. Single-cell analysis showed that the high expression of GSTP1 in prostate cancer epithelial cells was closely associated with the upregulation of the P53 pathway and apoptosis. Conclusions: Our study reveals that GSTP in Glutathione metabolism reduces the risk of prostate cancer and further analyzes the genetic association and mechanism of action between GSTP1 and prostate cancer.
Collapse
Affiliation(s)
- Shan Huang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
- Institute of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Hang Yin
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
- Institute of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
21
|
Jiang B, Bi Y, Chen Y, Bi J, Deng J, Zhang G. Case Report: Two cases of chemotherapy refractory aggressive variant prostate cancer with extreme durable response to PARP inhibitor. Front Oncol 2025; 15:1533627. [PMID: 40342821 PMCID: PMC12058769 DOI: 10.3389/fonc.2025.1533627] [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: 11/24/2024] [Accepted: 04/02/2025] [Indexed: 05/11/2025] Open
Abstract
Background Aggressive variant prostate cancer (AVPC) represents a distinct clinical subset characterized by resistance to novel hormone therapies and an unfavorable prognosis, frequently associated with the concurrent loss of tumor suppressor genes (TSG) such as PTEN, RB1, and TP53. While the progression-free survival (PFS) and overall survival (OS) of AVPC are relatively short, the optimal first-line treatment remains unclear. Presentation In this case report, we presented two de novo AVPC cases who have ultimately benefited from the usage of PARP inhibitors. The first patient was a 64-year-old male who was diagnosed during prostate biopsy featured by mutations in PTEN, and loss of RB1, BRCA2, ATM, and FANCA. He was treated with docetaxel/albumin-bound paclitaxel and cisplatin in the first line. Second-line therapy was applied with radiotherapy and Olaparib after failure of first-line therapy, resulting in a PSA response sustained for three years. The second case was a 75-year-old male with localized neuroendocrine feature and mutations in TP53, loss of RB1 and HDAC2. He was treated with sustained ADT and chemotherapy in the first-line treatment. Radiotherapy and Fluzoparib + abiraterone was applied as subsequent treatments with a PSA response for 2 years. Conclusions These two cases demonstrating a satisfactorily durable response to PARP inhibitors indicating its clinical benefit in AVPC population with detected DNA damage response (DDR) defects. The survival improvement with PARP inhibitors observed in our clinical experiences, along with current advances in tumor sequencing provide more information on future clinical trials and explorations of innovative therapies in AVPC population.
Collapse
Affiliation(s)
- Bohao Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yifan Bi
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiming Chen
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianbin Bi
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Deng
- Third Department of Medical Oncology, The Fifth People Hospital of Shenyang, Shenyang, Liaoning, China
| | - Gejun Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
22
|
Pan J, Zhu B, Wu J, Ni X, Li X, Jin S, Ma W, Liu X, Hu X, Gan H, Wang H, Wang B, Zhang Q, Song S, Liu C, Ye D, Zhu Y. Homologous recombination repair gene alteration is strongly associated with more prostate-specific membrane antigen-positive metastases in newly diagnosed hormone-sensitive prostate cancer with ≤5 conventional imaging defined distant metastases. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07278-z. [PMID: 40237795 DOI: 10.1007/s00259-025-07278-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The differences in imaging between prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) and conventional imaging (CI) significantly impact disease staging, subsequently influencing the scope and clinical efficacy of stereotactic body radiotherapy (SBRT). In this multicenter retrospective study, we aimed to explore the effect of homologous recombination repair (HRR) gene status on the imaging differences between PSMA PET/CT and CI, which may have important implications for treatment selection. PATIENTS AND METHODS A total of 1214 newly diagnosed hormone-sensitive prostate cancer (HSPC) patients with ≤ 5 CI-defined distant metastatic lesions were included. All patients underwent PSMA PET/CT, CI, and circulating tumor DNA testing for 19 HRR genes. The PSA response was defined as achieving a PSA level of < 0.1 ng/ml after 6 months. RESULTS The median PSA level was 22.7 ng/ml. In the comparison of bone metastasis detection rates, the proportion of patients with higher detection rates on PSMA PET/CT than on CI was similar between BRCA mutation carriers and those with other HRR gene mutations (43.6% vs. 39.3%, p = 0.554), yet significantly higher in both groups compared to non-mutation carriers (20.5%, p = 0.00001). Similar results were observed in the analysis of metastasis detection rates for distant lymph nodes and regional lymph nodes. In non-metastatic HSPC patients without PSMA-positive distant disease who accepted radical prostatectomy (RP), patients without HRR gene mutations exhibited a significantly higher PSA response rate compared to HRR gene mutated patients (96.9% vs. 90.2%, p = 0.003). CONCLUSIONS HRR gene alterations were significantly associated with a higher number of PSMA-positive metastases in newly diagnosed HSPC with ≤ 5 CI-defined distant metastases and worse outcomes in non-metastatic HSPC accepting RP. This finding suggests that HRR gene status should be considered as a potential indicator for recommending PSMA PET/CT in the design of clinical trials involving SBRT and in shaping imaging strategies.
Collapse
Affiliation(s)
- Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xudong Ni
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengming Jin
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiwei Ma
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaohang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaoxin Hu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hongkai Wang
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Beihe Wang
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20 dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Shaoli Song
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Chang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
- Shanghai Genitourinary Cancer Institute, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, People's Republic of China.
- Shanghai Genitourinary Cancer Institute, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Jansen JP, Brewer I, Flottemesch T, Grossman JP. The Health Inequality Impact of Darolutamide for Non-Metastatic Castration-Resistant Prostate Cancer in the United States: A Distributional Cost-Effectiveness Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025:S1098-3015(25)00123-8. [PMID: 40204257 DOI: 10.1016/j.jval.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
OBJECTIVES Non-Hispanic (NH) Black patients are disproportionally affected by nonmetastatic castration-resistant prostate cancer (nmCRPC). The objective was to quantify the health inequality impact of darolutamide + androgen deprivation therapy (ADT) relative to ADT for nmCRPC in the United States using a distributional cost-effectiveness analysis. METHODS With a health economic model, quality-adjusted life years (QALYs) and costs were estimated for NH-White, NH-Black, NH-Asian, and Hispanic patients. Given the lifetime risk of nmCRPC and assuming equally distributed opportunity costs, the incremental net health benefits of darolutamide were calculated, which were used to estimate general population quality-adjusted life expectancy at birth (QALE) by race and ethnicity with and without darolutamide. The extent of QALYs and QALE differences between race and ethnicity subgroups with each strategy was quantified with an inequality index, and their difference defined as the inequality impact of darolutamide. RESULTS Darolutamide + ADT resulted in an additional 1.04 (95% uncertainty interval 0.56-1.51) QALYs per treated patient relative to ADT, with the greatest gain observed among NH-Black patients (1.48 [0.48-2.71]). The relative inequality in QALYs among patients reduced by 66%, from an inequality score of 0.033 (0.004-0.082) with ADT to 0.011 (0.000-0.051) with darolutamide + ADT. Factoring in disease risk and health opportunity costs, nmCRPC treatment with darolutamide resulted in the largest net gain in QALYs among the NH-Black population, thereby having a favorable impact on inequalities in QALE. CONCLUSIONS Darolutamide + ADT results in greater and a more even distribution of QALYs than ADT for nmCRPC. The greatest gains among NH-Black individuals implies a favorable health inequality impact with darolutamide.
Collapse
Affiliation(s)
- Jeroen P Jansen
- Precision AQ, Health Economics and Outcomes Research, Bethesda, MD, USA.
| | - Iris Brewer
- Precision AQ, Health Economics and Outcomes Research, Bethesda, MD, USA
| | | | | |
Collapse
|
24
|
Smani S, DuBois J, Ajjawi I, Sohoni N, Choksi AU, Lokeshwar SD, Kim IY, Renzulli JF. Advances in Current Treatment Paradigms for Metastatic Hormone-Sensitive Prostate Cancer. J Clin Med 2025; 14:2565. [PMID: 40283395 PMCID: PMC12028174 DOI: 10.3390/jcm14082565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Metastatic hormone-sensitive prostate cancer (mHSPCa) presents de novo or represents significant disease progression and requires systemic treatment. However, progression to castration resistance is inevitable. The treatment landscape has evolved with the introduction of intensified systemic therapy, including androgen deprivation therapy (ADT) combined with either androgen receptor signaling inhibitors (ARSIs) or cytotoxic chemotherapy (doublet therapy) or combined therapy with both agents (triplet therapy). Landmark trials such as CHAARTED, STAMPEDE, LATITUDE, ENZAMET, and TITAN have established combination therapies as the standard of care, demonstrating significant overall survival benefits. More recently, triplet therapy-integrating ADT, docetaxel, and an ARSI-has emerged as an effective approach, particularly in high-volume metastatic disease, as supported by ARASENS and PEACE-1. Advances in imaging, such as PSMA PET-CT, have improved disease detection, allowing earlier detection of metastasis and appropriate therapy. Similarly, genomic profiling has enabled biomarker-driven, personalized treatment strategies. The role of treatment of the primary tumor, by either radiation therapy or cytoreductive prostatectomy, in low-volume disease continues to be explored. As novel therapies, targeted agents, and immunotherapies undergo investigation, optimizing treatment selection based on disease burden, molecular characteristics, and patient factors will be essential. The future of mHSPCa management lies in multidisciplinary, precision-based approaches to improve patient outcomes while balancing treatment efficacy and tolerability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Joseph F. Renzulli
- Department of Urology, Yale School of Medicine, New Haven, CT 06520, USA; (S.S.); (J.D.); (I.A.); (N.S.); (A.U.C.); (S.D.L.); (I.Y.K.)
| |
Collapse
|
25
|
Hammarlund N, Holt SK, Etzioni R, Morehead D, Lee JR, Wolff EM, Burrola-Mendez Y, Sage L, Gore JL, Nyame YA. The association of where patients with prostate cancer live and receive care on racial treatment inequities. J Natl Cancer Inst 2025; 117:713-718. [PMID: 39565920 PMCID: PMC11972675 DOI: 10.1093/jnci/djae302] [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: 08/21/2024] [Revised: 10/31/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Black individuals are less likely to be treated for prostate cancer even though they are more than twice as likely to die compared with White individuals. The complex causes of these inequities are influenced by social and structural factors, including racism, which contribute to the differential delivery of care. This study investigates how factors related to the location of where individuals live and receive care affect treatment inequities for prostate cancer between Black and White individuals. We hypothesize that both location and race independently influence treatment inequities. METHODS We used data from the Surveillance, Epidemiology, and End Results (SEER) cancer registry linked to Medicare claims to estimate the treatment inequity, as defined by differences in radiation or radical prostatectomy. Fixed effects at the physician, hospital, and patient ZIP code levels were incorporated to adjust for all time-invariant factors at these levels. RESULTS The results indicate that residential location-related factors explain only half of the treatment inequity, whereas provider- and hospital-level factors do not significantly account for disparities. Even after accounting for all time-invariant factors, significant differences in treatment rates persist. CONCLUSIONS The study highlights the importance of understanding race as a social construct and racism as a systemic and structural phenomenon in addressing treatment inequities. These findings provide a necessary step toward understanding equitable care and designing interventions to solve this inequity.
Collapse
Affiliation(s)
- Noah Hammarlund
- Department of Health Services Research Management & Policy, University of Florida, Gainesville, FL, United States
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
| | - Sarah K Holt
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Danté Morehead
- Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, WA, United States
| | - Jenney R Lee
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
| | - Erika M Wolff
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
| | - Yohali Burrola-Mendez
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
| | - Liz Sage
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
| | - John L Gore
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Yaw A Nyame
- Department of Urology, University of Washington Medical Center, Seattle, WA, United States
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
26
|
Viscuse P, Skelton WP, Devitt MM, Dreicer R. When You Get to the Fork in the Road, Take It: The Challenges in Managing Patients With Advanced Prostate Cancer. JCO Oncol Pract 2025; 21:467-475. [PMID: 39353159 DOI: 10.1200/op-24-00591] [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: 07/23/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
As is the case with most solid tumors, the heterogeneity of the disease biology of prostate cancer presents clinicians managing this disease with daily challenges. However, in contrast to other common cancers such as breast, lung, and colorectal cancers, there are unique challenges in prostate cancer management, including the variety of clinicians who manage aspects of the disease (urologists, medical oncologist, radiation oncologists) and the striking absence of prospective comparative data to inform the optimal sequence of systemic therapy in patients with metastatic castration-resistant disease. The purpose of this review is to attempt to assist practicing oncologists with sorting through the myriad of prostate cancer disease subsets and the challenges in making therapeutic decisions in multiple data-free zones given the absence of level 1 comparative clinical trials in the metastatic hormone-sensitive and castration-resistant states.
Collapse
Affiliation(s)
- Paul Viscuse
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| | - William P Skelton
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| | - Michael M Devitt
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| | - Robert Dreicer
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA
| |
Collapse
|
27
|
Egevad L, Micoli C, Delahunt B, Samaratunga H, Garmo H, Stattin P, Eklund M. Gleason scores provide more accurate prognostic information than grade groups. Pathology 2025; 57:293-296. [PMID: 39924438 DOI: 10.1016/j.pathol.2024.12.633] [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: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/30/2024] [Indexed: 02/11/2025]
Abstract
Prostate cancer grade is currently often reported both by Gleason scores and by grouping of the scores into five so-called International Society of Urological Pathology (ISUP) grades (also known as grade groups). Using population-based registry data from 172,112 men diagnosed with prostate cancer on needle biopsy, we recently investigated the outcome of Gleason score 8-10 prostate cancer with death due to prostate cancer and death from any cause as endpoints. There was a prognostic heterogeneity between Gleason scores 3+5, 4+4 and 5+3 (ISUP grade 4) and between Gleason scores 4+5, 5+4 and 5+5 (ISUP grade 5). This heterogeneity was lost when the grades collapsed into ISUP grades 4 and 5, respectively. On the other hand, there was also a prognostic overlap between these ISUP grades. The outcome of Gleason score 5+3 and 4+5 cancers was very similar. The prostate-specific mortality of Gleason scores 5+3 and 4+5 was 0.32 (95% confidence interval 0.27-0.36) and 0.30 (0.29-0.31), respectively, after 5 years and 0.44 (0.39-0.49) and 0.45 (0.44-0.46), respectively, after 10 years. The findings emphasise the importance of reporting the Gleason grades and scores for more accurate prognostic information of highly heterogeneous high-grade prostate cancers. It also questions the clinical value of the current recommendations of grouping of Gleason scores into ISUP grades or grade groups.
Collapse
Affiliation(s)
- Lars Egevad
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Chiara Micoli
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Brett Delahunt
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Hans Garmo
- Regional Cancer Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Pär Stattin
- Regional Cancer Centre, Uppsala University Hospital, Uppsala, Sweden; Department of Surgical Sciences Uppsala University, Uppsala, Sweden
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Aggarwal A, Bharadwaj S, Corredor G, Pathak T, Badve S, Madabhushi A. Artificial intelligence in digital pathology - time for a reality check. Nat Rev Clin Oncol 2025; 22:283-291. [PMID: 39934323 DOI: 10.1038/s41571-025-00991-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
The past decade has seen the introduction of artificial intelligence (AI)-based approaches aimed at optimizing several workflows across many medical specialties. In clinical oncology, the most promising applications include those involving image analysis, such as digital pathology. In this Perspective, we provide a comprehensive examination of the developments in AI in digital pathology between 2019 and 2024. We evaluate the current landscape from the lens of technological innovations, regulatory trends, deployment and implementation, reimbursement and commercial implications. We assess the technological advances that have driven improvements in AI, enabling more robust and scalable solutions for digital pathology. We also examine regulatory developments, in particular those affecting in-house devices and laboratory-developed tests, which are shaping the landscape of AI-based tools in digital pathology. Finally, we discuss the role of reimbursement frameworks and commercial investment in the clinical adoption of AI-based technologies. In this Perspective, we highlight both the progress and challenges in AI-driven digital pathology over the past 5 years, outlining the path forward for its adoption into routine practice in clinical oncology.
Collapse
Affiliation(s)
- Arpit Aggarwal
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Satvika Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Germán Corredor
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA
| | - Tilak Pathak
- Department of Biomedical Engineering, Emory University, Atlanta, GA, USA
| | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
29
|
Kishan AU, Sun Y, Tree AC, Hall E, Dearnaley D, Catton CN, Lukka HR, Pond G, Lee WR, Sandler HM, Feng FY, Nguyen PL, Incrocci L, Heemsbergen W, Pos FJ, Horwitz E, Wong JK, Hoffman KE, Hassanzadeh C, Kuban DA, Arcangeli S, Sanguineti G, Supiot S, Crehange G, Latorzeff I, Kalbasi TR, Steinberg ML, Valle LF, Loblaw A, Nikitas J, Roy S, Zaorsky NG, Jia AY, Spratt DE. Hypofractionated radiotherapy for prostate cancer (HYDRA): an individual patient data meta-analysis of randomised trials in the MARCAP consortium. Lancet Oncol 2025; 26:459-469. [PMID: 40112848 DOI: 10.1016/s1470-2045(25)00034-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Trials comparing moderately hypofractionated radiotherapy (MHFRT) to conventionally-fractionated radiotherapy (CFRT) for prostate cancer have varied considerably in intent (non-inferiority vs superiority) and MHFRT dose. We compare the efficacy and toxicity profiles of isodose MHFRT and dose-escalated MHFRT. METHODS This was an individual patient data meta-analysis that identified randomised phase 3 trials of CFRT versus MHFRT that had published individual patient-level data on efficacy and late toxicity. A systematic literature search using MEDLINE, Embase, trial registries, the Web of Science, Scopus, and relevant conference proceedings was initially conducted on Dec 15, 2023, and was re-conducted on Jan 8, 2025. Trials that did not publish efficacy data, did not publish late toxicity data, or did not use modern dose radiotherapy (≥70 Gy in 2 Gy equivalents) in the CFRT group were excluded. Individual patient data were provided to MARCAP by study investigators. Three separate meta-analyses were designed to compare efficacy (primary endpoint was progression-free survival), physician-scored late toxicity (co-primary endpoints were late grade 2 or higher genitourinary and late grade 2 or higher gastrointestinal toxic effects), and patient-reported outcomes (co-primary endpoints were clinically-significant decrements in patient-reported urinary or bowel quality of life) between patients receiving CFRT versus MHFRT. FINDINGS We identified 1696 records for review. Seven phase 3 trials comparing MHFRT with CFRT were eligible for inclusion in our analysis. Individual patient data were obtained from these seven studies (3454 patients from three trials comparing CFRT with isodose MHFRT and 2426 patients from four trials comparing CFRT with dose-escalated MHFRT). At a median follow-up of 5·4 years (IQR 4·6-7·2) for isodose MHFRT and 7·1 years (5·7-8·4) for dose-escalated MHFRT, no differences in progression-free survival were detected (hazard ratio 0·92, 95% CI 0·81-1·05; p=0·21 and 0·94, 0·82-1·09; p=0·43 respectively). No increased odds of grade 2 or higher genitourinary toxic effects were identified for either isodose (odds ratio [OR] 1·16, 95 CI% 0·86-1·57; p=0·32) or dose-escalated MHFRT (1·20, 0·95-1·51; p=0·13). The odds of grade 2 or higher gastrointestinal toxic effects were significantly higher with dose-escalated (OR 1·48, 95% CI 1·14-1·92; p=0·0035) but not isodose MHFRT (1·30, 0·59-2·87; p=0·51). Isodose MHFRT was not found to show different odds of urinary quality-of-life decrement (OR 1·03, 95% CI 0·51-2·09; p=0·93) or bowel quality-of-life decrement (0·76, 0·40-1·43; p=0·39). Dose-escalated MHFRT was associated with greater odds of bowel quality-of-life decrement (OR 1·68, 95% CI 1·07-2·61; p=0·023), but no evidence of greater urinary quality-of-life decrement was found (1·57, 0·87-2·85; p=0·13). INTERPRETATION Isodose MHFRT and dose-escalated MHFRT both have similar efficacy compared with CFRT, but dose-escalated MHFRT is associated with higher physician-scored and patient-reported bowel toxicity. Isodose regimens, eg, 60 Gy in 20 fractions, should be the standard MHFRT regimen for localised prostate cancer. FUNDING None.
Collapse
Affiliation(s)
- Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA.
| | - Yilun Sun
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alison C Tree
- Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Emma Hall
- Institute of Cancer Research, London, UK
| | - David Dearnaley
- Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Charles N Catton
- Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Himanshu R Lukka
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, ON, Canada
| | | | - W Robert Lee
- Department of Radiation Oncology, Duke University, Durham, NC, USA
| | - Howard M Sandler
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Paul L Nguyen
- Department of Radiation Oncology, Dana- Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA, USA
| | - Luca Incrocci
- Department of Radiotherapy, Erasmus Medical Center, Rotterdam, Netherlands
| | - Wilma Heemsbergen
- Department of Radiotherapy, Erasmus Medical Center, Rotterdam, Netherlands
| | - Floris J Pos
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Eric Horwitz
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Jessica Karen Wong
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Karen E Hoffman
- Division of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Comron Hassanzadeh
- Division of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Deborah A Kuban
- Division of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stefano Arcangeli
- Department of Medicine and Surgery, University of Milan Bicocca, Milan, Italy
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Stephane Supiot
- Institut de Cancérologie de l'Ouest, Saint Herblain, Nantes, France
| | - Gilles Crehange
- Department of Radiation Oncology, Institut Curie, Saint-Cloud, France
| | - Igor Latorzeff
- Department of Radiation Oncology, Oncorad Clinique Pasteur, Toulouse, France
| | - Tahmineh Romero Kalbasi
- Department of Medicine Statistical Core, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael L Steinberg
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Luca F Valle
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA; Greater Los Angeles VA Medical Center, Los Angeles, CA, USA
| | - Andrew Loblaw
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - John Nikitas
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Soumyajit Roy
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Angela Y Jia
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
30
|
Uslu H, Şahin D, İbişoğlu E, Tatoğlu MT. PRIMARY scoring in 68Ga-PSMA PET/CT: correlation with prostate cancer risk groups and its potential impact on active surveillance. Ann Nucl Med 2025; 39:334-341. [PMID: 39579268 DOI: 10.1007/s12149-024-02004-5] [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: 08/22/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
OBJECTIVE The PRIMARY scoring system is a scale designed to identify clinically significant intraprostatic malignancies on 68Ga-PSMA PET/CT images. Active surveillance is a management method for patients with low-risk prostate cancer. In this study, we aimed to assess the efficacy of PRIMARY scoring in identifying appropriate candidates for active surveillance based on the distribution within prostate cancer risk groups. METHODS The data of 134 patients diagnosed with PCa by biopsy who underwent 68Ga-PSMA PET/CT imaging for post-diagnostic staging purposes were retrospectively analyzed. Age, total PSA, ISUP grade, prostate lesion SUVmax values, PI-RADS scores, and PRIMARY scores were recorded. Patients were classified into low-risk and intermediate/high-risk groups. RESULTS In the intermediate/high-risk group, the PRIMARY score was 1-2 in 17.6% and 3-5 in 82.4% of patients. In the low-risk group, the PRIMARY score was 1-2 in 34.7% and 3-5 in 65.3% of patients. None of the patients in the low-risk group had a PRIMARY score of 5. The most frequent PRIMARY score in both groups was 4, and there was a significant difference between the average SUVmax values of the intermediate/high and low-risk groups with a PRIMARY score of 4 (p = 0.018). The sensitivity of PRIMARY scoring in detecting patients in the intermediate/high-risk group was 82.3%, the specificity was 34.6%, and the positive predictive value (PPV) was 68.6%. When a cut-off SUVmax value 5.0 was used for the PRIMARY score of 4, the sensitivity was 67.0%, the specificity was 65.3% and the PPV was 77.0%. In the ROC analysis, the area under the curve was 0.727 for PRIMARY scoring, 0.662 for PI-RADS, and 0.744 for their combined mean. CONCLUSION The PRIMARY scoring system can complement PI-RADS scoring in mpMRI for selecting patients suitable for active surveillance. Revising the PRIMARY score 4 with an SUVmax cut-off value may increase the specificity.
Collapse
Affiliation(s)
- Hatice Uslu
- Clinic of Nuclear Medicine, Istanbul Medeniyet University Göztepe Prof. Dr. Süleyman Yalçın City Hospital, Eğitim Mah. Fahrettin Kerim Gökay Cd., 34722, Kadikoy, Istanbul, Turkey
| | - Dilruba Şahin
- Clinic of Nuclear Medicine, Istanbul Medeniyet University Göztepe Prof. Dr. Süleyman Yalçın City Hospital, Eğitim Mah. Fahrettin Kerim Gökay Cd., 34722, Kadikoy, Istanbul, Turkey.
| | - Ebru İbişoğlu
- Clinic of Nuclear Medicine, Istanbul Medeniyet University Göztepe Prof. Dr. Süleyman Yalçın City Hospital, Eğitim Mah. Fahrettin Kerim Gökay Cd., 34722, Kadikoy, Istanbul, Turkey
| | - Mehmet Tarık Tatoğlu
- Clinic of Nuclear Medicine, Istanbul Medeniyet University Göztepe Prof. Dr. Süleyman Yalçın City Hospital, Eğitim Mah. Fahrettin Kerim Gökay Cd., 34722, Kadikoy, Istanbul, Turkey
| |
Collapse
|
31
|
Wang JH, Shi X, Tran PT, Sutera P. Integrating Prostate Specific Membrane Antigen-PET into Clinical Practice for Prostate Cancer. PET Clin 2025; 20:205-217. [PMID: 39924369 PMCID: PMC12012819 DOI: 10.1016/j.cpet.2025.01.001] [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] [Indexed: 02/11/2025]
Abstract
Prostate surface membrane antigen (PSMA)-PET imaging has significantly shaped the clinical management of prostate cancer, from localized to metastatic disease. It outperforms conventional imaging in both primary staging and detecting recurrence. PSMA-PET incorporation into the clinical workflow can alter treatment decisions, though the impact of observed stage migration on patient outcomes has yet to be well-characterized. There is growing interest in using PSMA-PET to predict treatment response across all stages of prostate cancer, and to select patients for PSMA radioligand therapy. Use of PSMA-PET will continue to expand for clinical applications as its role becomes better defined through prospective studies.
Collapse
Affiliation(s)
- Jarey H Wang
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, 401 N Broadway Street, Baltimore, MD 21287, USA
| | - Xiaolei Shi
- Department of Hematology/Oncology, University of Maryland Medical Center, 22 S. Greene Street, Baltimore, MD 21201, USA
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland Medical Center, 850 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Philip Sutera
- Department of Radiation Oncology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| |
Collapse
|
32
|
Patel S, Zhu K, Dave CV, Ghajar M, Zhang Y, Saraiya B, Bandera EV, Khosrow-Khavar F. Comparative Cardiovascular Safety of Gonadotropin-releasing Hormone Antagonists and Agonists Among Patients Diagnosed with Prostate Cancer: A Systematic Review and Meta-analysis of Real-world Evidence Studies. Eur Urol Oncol 2025; 8:510-519. [PMID: 39343637 DOI: 10.1016/j.euo.2024.09.004] [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: 05/03/2024] [Revised: 08/11/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND AND OBJECTIVE Gonadotropin-releasing hormone (GnRH) antagonists and agonists are cornerstone treatments in prostate cancer. However, evidence regarding the comparative cardiovascular safety of these drugs from clinical trials is inconclusive. The objective of this study was to systematically assess the risk of adverse cardiovascular events of GnRH antagonists compared with GnRH agonists across real-world evidence studies. METHODS We conducted a systematic search of PubMed, Embase, Cochrane Library, Scopus, and Web of Science (2008-2023). We included real-world evidence studies comparing the risk of cardiovascular outcomes of GnRH antagonists with those of GnRH agonists among patients with prostate cancer. We conducted a meta-analysis of effect estimates across studies at a low or moderate risk of bias, assessed via the Risk of Bias in Non-Randomized Studies of Interventions (ROBINS-I) tool, using random-effect models. KEY FINDINGS AND LIMITATIONS Among ten included studies, four were classified as having a moderate and six as having a serious risk of bias. Across three studies at a moderate risk of bias in the primary analysis, degarelix was associated with an increased risk (pooled relative risk [RR]: 1.31, 95% confidence interval [CI]: 1.14-1.51) of major adverse cardiovascular events (MACEs). An augmented risk was observed in two studies among patients with a history of cardiovascular disease (pooled RR: 1.31, 95% CI: 1.11-1.56) compared with one study among patients without a history of cardiovascular disease (RR: 1.15, 95% CI: 0.83-1.59). CONCLUSIONS AND CLINICAL IMPLICATIONS Real-world evidence studies indicate that degarelix, compared with GnRH agonists, is associated with a modest increased risk of MACEs, particularly among patients with a history of cardiovascular disease. However, residual confounding due to the treatment of high-risk patients with degarelix may account for these findings. Additional large studies with detailed data on tumor characteristics and cardiovascular risk factors are needed to confirm these findings. PATIENT SUMMARY In this systematic evaluation of evidence among patients diagnosed with prostate cancer in routine care, degarelix was associated with higher cardiovascular adverse outcomes than gonadotropin-releasing hormone agonists.
Collapse
Affiliation(s)
- Savan Patel
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Department of Biostatistics and Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Kexin Zhu
- Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Healthcare Policy and Aging Research, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Chintan V Dave
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Healthcare Policy and Aging Research, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Mina Ghajar
- Health Sciences Libraries, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Yingting Zhang
- Health Sciences Libraries, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | | | - Elisa V Bandera
- Department of Biostatistics and Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Rutgers Cancer Institute, New Brunswick, NJ, USA
| | - Farzin Khosrow-Khavar
- Department of Biostatistics and Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Healthcare Policy and Aging Research, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Rutgers Cancer Institute, New Brunswick, NJ, USA.
| |
Collapse
|
33
|
Tosoian JJ. Active surveillance for prostate cancer: How active is too active? J Natl Cancer Inst 2025; 117:583-585. [PMID: 39868757 DOI: 10.1093/jnci/djae342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/28/2025] Open
Affiliation(s)
- Jeffrey J Tosoian
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Nashville, TN, United States
| |
Collapse
|
34
|
Hoeh B, Preisser F, Zattoni F, Kretschmer A, Westhofen T, Olivier J, Soeterik TFW, van den Bergh RCN, Mandel P, Graefen M, Tilki D. Risk of Biochemical Recurrence and Metastasis in Prostate Cancer Patients Treated with Radical Prostatectomy After a 10-year Disease-free Interval. Eur Urol Oncol 2025; 8:372-379. [PMID: 39306583 DOI: 10.1016/j.euo.2024.08.008] [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: 08/02/2024] [Accepted: 08/27/2024] [Indexed: 03/24/2025]
Abstract
BACKGROUND AND OBJECTIVE Prostate-specific antigen (PSA) testing is used to follow up prostate cancer (PCa) patients treated with radical prostatectomy (RP). Research on PSA thresholds for identifying PCa patients with biochemical recurrence (BCR) who are at a higher risk of progression yielded inconclusive results. This study aims to investigate the risk of late BCR in PCa patients treated with RP and long postoperative (120 mo) undetectable PSA follow-up, and to identify prognostic factors for late BCR within this patient cohort. METHODS PCa patients treated with curative RP (1992-2012) and free of BCR during the first 120 mo following RP were retrospectively identified within five European tertiary centers; BCR was defined as two consecutive PSA values of ≥0.2 ng/ml. Kaplan-Meier and Cox regression models tested for an association between BCR and patient or tumor characteristics. KEY FINDINGS AND LIMITATIONS The study cohort consisted of 4639 patients, of whom 243 (5.2%) developed BCR at a medium follow-up of 147 mo. Of those with BCR, 23 (9.5%) subsequently developed metastatic progression. In Kaplan-Meier models, BCR-free survival differed according to advanced tumor status. In multivariable Cox regression models, pT stage (pT3a: hazard ratio [HR]: 1.46; pT3b: HR: 2.42), pathological Gleason score (pGS 3 + 4: HR: 1.71; pGS ≥4 + 3: HR: 2.47), surgical margin (R1/Rx: HR: 1.72), and pNx stage (pNx: HR: 0.72) represented independent predictors for BCR (all p < 0.05). Conversely, age, PSA at diagnosis, and year of surgery failed to achieve independent predictor status for BCR. CONCLUSIONS AND CLINICAL IMPLICATIONS Among PCa patients with an uneventful follow-up of at least 10 yr after RP, still one in 20 patients subsequently develop late BCR. Nevertheless, late BCR and subsequent progression to metastasis (0.3%) rates in patients with pT2 stage and pGS ≤3 + 4 were strikingly low, implicating that abandoning follow-up beyond an uneventful period of 10 yr is justifiable within this cohort of patients. PATIENT SUMMARY In this study, prostate cancer patients treated with a radical prostatectomy and at least 10 yr of uneventful prostate-specific antigen testing were identified within five European centers. Relying on these patients, the rate of subsequent late biochemical recurrence was calculated and risk factors were identified for biochemical recurrence following 10 yr of uneventful prostate-specific antigen testing.
Collapse
Affiliation(s)
- Benedikt Hoeh
- Department of Urology, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Felix Preisser
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Fabio Zattoni
- Urologic Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy; Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Alexander Kretschmer
- Department of Urology, University Hospital Munich, Campus Großhadern, Ludwig-Maximilians University, Munich, Germany
| | - Thilo Westhofen
- Department of Urology, University Hospital Munich, Campus Großhadern, Ludwig-Maximilians University, Munich, Germany
| | | | - Timo F W Soeterik
- Department of Urology, St. Antonius Hospital, Nieuwegein, Utrecht, The Netherlands
| | | | - Philipp Mandel
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Graefen
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey.
| |
Collapse
|
35
|
Kato M, Sato H, Naito Y, Yamamoto A, Kawanishi H, Nakano Y, Nishikimi T, Kobayashi M, Kondo A, Hirabayashi H, Katsuno S, Sakamoto F, Kimura T, Yamamoto S, Araki H, Tochigi K, Ito F, Hatsuse H, Sassa N, Hirakawa A, Akamatsu S, Tsuzuki T. Prospective observational study on the relationships between genetic alterations and survival in Japanese patients with metastatic castration-sensitive prostate cancer: the impact of IDC-P. Int J Clin Oncol 2025; 30:789-796. [PMID: 39937427 PMCID: PMC11947062 DOI: 10.1007/s10147-025-02707-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Intraductal Carcinoma of the Prostate (IDC-P) is a significant prognostic indicator for prostate cancer, which demonstrates significant associations with homologous recombination repair gene mutations (HRRm) and alterations in tumor suppressor genes. However, no study in Japan has investigated the association between IDC-P and genetic mutations in men with metastatic castration-sensitive prostate cancer (mCSPC). METHODS This prospective observational study enrolled 102 de novo mCSPC (LATITUDE high-risk) patients diagnosed between 2018 and 2021, with subsequent monitoring of survival outcomes. A single genitourinary pathologist evaluated all needle biopsy slides. Genetic analyses were performed using the Myriad myChoice HRD plus™. These genetic analyses covered 108 genetic loci, including 15 HRRm genes, with a success rate of 91%. RESULTS Genetic alterations were observed in 79 patients (77.5%), with 20 exhibiting HRRm (19.6%). Common genetic alterations included FOXA1 (29.4%) and TP53 (17.6%) mutations; BRCA (9.8%) mutations were the most frequent HRRm (BRCA1:2 cases, BRCA2:8 cases, including 6 biallelic). IDC-P-positive patients demonstrated a significantly higher frequency of genetic aberrations (82.6% vs. 50%, p = 0.0082). Patients with biallelic BRCA2, TP53, and PTEN mutations exhibited significantly poorer cancer-specific survival. Multivariate analysis identified lactate dehydrogenase (LDH) (HR 1.005, p = 0.035), TP53 mutations (HR 5.196, p < 0.001), biallelic BRCA2 mutations (HR 10.686, p = 0.005), and IDC-P as independent predictors of poor cancer-specific survival. No cancer-related deaths occurred in IDC-P-negative cases. CONCLUSION Our study emphasizes the significant association between IDC-P and an elevated incidence of genetic alterations in Japanese mCSPC patients, emphasizing the need for early genetic testing to guide therapeutic decision-making.
Collapse
Affiliation(s)
- Masashi Kato
- Department of Urology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, 3-35 Michishita-Cho, Nakamura-Ku, Nagoya, 453-8511, Japan.
| | - Hiroyuki Sato
- Division of Biostatistics and Data Science, Clinical Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yushi Naito
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akiyuki Yamamoto
- Department of Urology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | | | - Yojiro Nakano
- Department of Urology, Tosei General Hospital, Seto, Japan
| | - Toshinori Nishikimi
- Department of Urology, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | | | - Atsuya Kondo
- Department of Urology, Kariya Toyota General Hospital, Kariya, Japan
| | - Hiroki Hirabayashi
- Department of Urology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, 3-35 Michishita-Cho, Nakamura-Ku, Nagoya, 453-8511, Japan
| | - Satoshi Katsuno
- Department of Urology, Okazaki City Hospital, Okazaki, Japan
| | | | - Tohru Kimura
- Department of Urology, Japan Community Healthcare Organization Chukyo Hospital, Nagoya, Japan
| | | | | | - Kosuke Tochigi
- Department of Urology, Yokkaichi Municipal Hospital, Yokkaichi, Japan
| | - Fumihiro Ito
- Department of Urology, Gifu Prefectural Tajimi Hospital, Tajimi, Japan
| | - Hatsuro Hatsuse
- Department of Urology, Ichinomiya Municipal Hospital, Ichinomiya, Japan
| | - Naoto Sassa
- Department of Urology, Aichi Medical University, Nagakute, Japan
| | - Akihiro Hirakawa
- Division of Biostatistics and Data Science, Clinical Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shusuke Akamatsu
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| |
Collapse
|
36
|
Tao XY, Li QQ, Dong SS, Wang H, Yang YQ, Yang X, Zeng Y. Long noncoding HOXD-AS1: a crucial regulator of malignancy. Front Cell Dev Biol 2025; 13:1543915. [PMID: 40206400 PMCID: PMC11979210 DOI: 10.3389/fcell.2025.1543915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) play a crucial role in the occurrence and progression of various cancers. HOXD-AS1, an antisense RNA 1 of the lncRNA HOXD cluster, (also known as HAGLR, MIR7704HG, Mdgt, and STEEL), is located at human chromosome 2q31.1. Recent studies have demonstrated that the abnormal expression of HOXD-AS1 is significantly correlated with the clinicopathological features of patients with various tumors. The expression of HOXD-AS1 is abnormal in various tumors, affecting tumor cell proliferation, apoptosis, metastasis, invasion, metabolism, and drug resistance. HOXD-AS1 is important for cancer diagnosis and prognosis evaluation. Detecting its expression level helps judge cancer progression and predict patient survival. It is a therapeutic target and biomarker for early diagnosis and prognosis, with good clinical application prospects. This article reviews the role, molecular mechanisms, and potential clinical value of HOXD-AS1 in malignant tumor development.
Collapse
Affiliation(s)
- Xiang-Yuan Tao
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qian-Qian Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shan-Shan Dong
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui Wang
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yu-Qing Yang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xi Yang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yong Zeng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
37
|
Jiang B, Wang B, Chen Y, Chen Y, Li B, Bi J. Comparative therapeutic efficacy and safety of first-line and second-line therapies for metastatic castration-resistant prostate cancer: a systematic review and network meta-analysis. EClinicalMedicine 2025; 81:103129. [PMID: 40104085 PMCID: PMC11914769 DOI: 10.1016/j.eclinm.2025.103129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Background There is no cross-sectional comparison on therapeutic and adverse effects for treatments of metastatic castration-resistant prostate cancer (mCRPCa). We aimed to horizontally compare them for all common first-line and second-line therapies. Methods We conducted a network meta-analysis with a systematic review in four databases (Pubmed, Web of Science, Embase, and Cochrane Library) up to January 5th, 2025. All randomized controlled trials (RCT) related to mCRPCa treatments with a clear description in study design were included. Endpoints included the radiographic progression-free survival (rPFS), overall survival (OS), time to PSA progression (TTPP), PSA progression rate (PSARR), and adverse events. All data was extracted by two researchers and analyzed with Gemtc package in R and Stata15. This NMA protocol was registered online (ID: CRD42025633178). Findings After screening among 33,694 articles, 24 RCTs involving 13,059 cases were included. For first-line treatments, combination therapies with second-generation androgen receptor inhibitors (ARIs) showed superior efficacies in OS [HR of poly(ADP-ribose) polymerase inhibitors (PARPi) + ARI: 0.63 (0.32,1.25)], TTPP [HR of Lu177 + ARI: 0.07 (0.01,0.87)] and PSARR [RR of Lu177 + ARI: 33.02 (15.56,71.62)] with the highest SUCRA (Surface under the Cumulative Ranking Curve) (72%, 91% and 97% respectively). PARPi + ARI also performed best for rPFS (SUCRA: 85%, with an insignificant HR [0.12 (0.02,2.35)]. For post-docetaxel second-line treatments, ARI also emerged as the preferred option. Efficacies of post-ARI second-line treatments were not evaluated due to the lack of related RCTs. No obvious heterogeneity and publication bias was detected during the therapeutic comparison. Interpretation This study provided comparative evidence for first-line and post-chemotherapy second-line mCRPCa treatment options. Second-generation ARIs exhibited good efficacy, particularly when combined with other treatments. However, the safety analysis necessitated balance between benefits and adverse events, especially for combination therapies. Stronger evidence is needed through direct comparisons in future clinical trials. Funding The study was supported by The National Natural Science Foundation of China (No. 82172568).
Collapse
Affiliation(s)
- Bohao Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Benqiao Wang
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Yiming Chen
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Yaang Chen
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Bohan Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Jianbin Bi
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| |
Collapse
|
38
|
Zhou Z, Li T, Zhang Y, Zhou X, Song X, Ji S, Huang Y, Zhang Y, Ruan Y. PCBP2 promotes immune evasion via cGAS-STING pathway in biochemical recurrence of prostate cancer. APL Bioeng 2025; 9:016112. [PMID: 40051782 PMCID: PMC11884866 DOI: 10.1063/5.0250173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/22/2025] [Indexed: 03/09/2025] Open
Abstract
Immunotherapy resistance is a significant obstacle in the treatment of prostate cancer (PCa), primarily due to immune evasion mechanisms. This study aims to explore cancer-intrinsic immune evasion-related genes (CIERGs) in PCa and develop a predictive signature for biochemical recurrence (BCR). Bulk RNA-seq data and single-cell RNA-sequencing (scRNA-seq) were obtained from TCGA and Gene Expression Omnibus database. The scRNA-seq data analysis revealed higher immune evasion scores in tumor cells compared to normal cells. Differentially expressed genes from TCGA-PRAD and GSE70769 cohorts were intersected with 182 core immune evasion genes, followed by univariate Cox regression, identifying 48 CIERGs significantly associated with BCR. Nonnegative matrix factorization (NMF) clustering revealed two immune evasion-related PCa subtypes. A risk signature based on CIERGs was developed using LASSO regression, and a nomogram was created to predict BCR-free survival. Among the 48 identified CIERGs, poly(C)-binding protein 2 (PCBP2) emerged as a key risk factor associated with poor prognosis in PCa, and its function was validated in vitro. NMF clustering identified two subtypes, with the C1 subtype having a poorer prognosis. Gene Set Variation Analysis highlighted enrichment in cell cycle, extracellular matrix receptor interaction, and transforming growth factor-beta signaling pathways in the C1 subtype. A CIERGs-based risk signature, including six key genes, was developed and validated, with the nomogram showing high predictive accuracy. In vitro experiments showed PCBP2 promotes PCa cell proliferation, migration, and invasion by inhibiting the cyclic GMP-AMP synthase-STING pathway. The CIERGs signature provides a precise prediction of BCR, with PCBP2 emerging as a potential therapeutic target due to its inhibition of the cGAS-STING pathway in PCa.
Collapse
Affiliation(s)
- Zeng Zhou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiewen Li
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichen Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuehao Zhou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Song
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiyu Ji
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yishu Huang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Ruan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Guglielmo P, Buffi N, Porreca A, Setti L, Aricò D, Muraglia L, Evangelista L. Current insights on PSMA PET/CT in intermediate-risk prostate cancer: a literature review. Ann Nucl Med 2025; 39:247-254. [PMID: 39812950 DOI: 10.1007/s12149-025-02015-w] [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: 07/13/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
The purpose of this systematic review was to evaluate the role of PSMA PET/CT in intermediate-risk prostate cancer (PCa) patients, to determine whether it could help improve treatment strategy and prognostic stratification. A systematic literature search up to May 2024 was conducted in the PubMed, Embase and Scopus databases. Articles with mixed risk patient populations, review articles, editorials, letters, comments, or case reports were excluded. The quality of the papers was assessed by using the CASP criteria. The literature search returned 1111 studies; however, 1105 articles were excluded, and therefore 6 full-text papers were retrieved for the final analysis. Three out of six papers focused on the utility of SUVmax in identifying high ISUP grade in patients with intermediate-risk PCa. The latest three papers discussed the controversial role of PSMA PET/CT in predicting the lymph node involvement, mainly in the case of favorable subset. PSMA PET has completely changed the management of patients with PCa; indeed its role is still undefined in patients with intermediate-risk disease. Future perspective is to investigate larger cohorts of intermediate-risk PCa patients, to fully recognize the added value offered by PSMA PET in this category of subjects.
Collapse
Affiliation(s)
- Priscilla Guglielmo
- Nuclear Medicine Unit, Humanitas Gavazzeni, Bergamo, Italy.
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
| | - Nicolò Buffi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Urology, Humanitas Clinical and Research Institute IRCCS, Rozzano, Italy
| | - Angelo Porreca
- Department of Oncological Urology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Lucia Setti
- Nuclear Medicine Unit, Humanitas Gavazzeni, Bergamo, Italy
| | - Demetrio Aricò
- Department of Nuclear Medicine, Humanitas Oncological Centre of Catania, 95125, Catania, Italy
| | - Lorenzo Muraglia
- Nuclear Medicine Unit, Humanitas Clinical and Research Institute IRCCS, Rozzano, Italy
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Nuclear Medicine Unit, Humanitas Clinical and Research Institute IRCCS, Rozzano, Italy
| |
Collapse
|
40
|
Ding CKC, Greenland NY, Sirohi D, Lotan TL. Molecular Landscape of Aggressive Histologic Subtypes of Localized Prostate Cancer. Surg Pathol Clin 2025; 18:1-12. [PMID: 39890297 DOI: 10.1016/j.path.2024.10.001] [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: 02/03/2025]
Abstract
Despite incredible progress in describing the molecular underpinnings of prostate cancer over the last decades, pathologic examination remains indispensable for predicting aggressive behavior in the localized setting. Beyond pathologic grade, specific histologic findings have emerged as critical prognostic or predictive indicators. Here, the authors review molecular correlates of aggressive histologic subtypes of prostate cancer in the localized setting, demonstrating that many of the signature molecular alterations found in metastatic disease-such as tumor suppressor gene loss and DNA repair defects-are enriched in primary disease with adverse histologic features, presaging aggressive behavior, and presenting opportunities for earlier germline screening or targeted therapies.
Collapse
Affiliation(s)
- Chien-Kuang C Ding
- Department of Pathology, University of California, San Francisco (UCSF), 1825 4th Street, M2370, San Francisco, CA 94158, USA
| | - Nancy Y Greenland
- Department of Pathology, University of California, San Francisco (UCSF), 1825 4th Street, M2370, San Francisco, CA 94158, USA
| | - Deepika Sirohi
- Department of Pathology, University of California, San Francisco (UCSF), 1825 4th Street, M2370, San Francisco, CA 94158, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
41
|
Fanelli GN, Nuzzo PV, Pederzoli F, Loda M. Deciphering Complexity: The Molecular Landscape of Castration-Resistant Prostate Cancer. Surg Pathol Clin 2025; 18:25-39. [PMID: 39890307 PMCID: PMC11787547 DOI: 10.1016/j.path.2024.10.003] [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] [Indexed: 02/03/2025]
Abstract
Despite improvements in diagnosis and treatment approaches, prostate cancer (PC) remains a leading cause of cancer-related death in men. PC progresses through various stages, mostly driven by androgen receptor signaling. However, after androgen deprivation therapies, in a significant portion of patients, several different molecular mechanisms contribute to the development of castration resistance. Delving deeply into the molecular landscape of castration-resistant PC, grasping the selective pressures exerted by therapies, and understanding the drivers of lineage plasticity is pivotal to prevent progression. Targeting genetic and epigenetic alterations that drive this transition will guide clinical management strategies and possibly prevent and/or treat lethal disease.
Collapse
Affiliation(s)
- Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, Pisa 56125, Italy
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
42
|
Bolek H, Yazgan SC, Ceylan F, Esteban-Villarrubia J, Arslan C, Kuş T, Tural D, Sendur MAN, Kucuk NO, Çıngı Özdemir E, Castro E, Yekedüz E, Ürün Y. Comparison of two alternative sequences with cabazitaxel and 177Lu-PSMA-617 in metastatic castration-resistant prostate cancer: A retrospective multicenter study (LuCaS). Eur J Cancer 2025; 217:115226. [PMID: 39813762 DOI: 10.1016/j.ejca.2025.115226] [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: 11/01/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Cabazitaxel and 177Lu-PSMA-617 have been shown to improve survival in patients with metastatic castration-resistant prostate cancer (mCRPC) previously treated with docetaxel and androgen receptor pathway inhibitors (ARPI). we aimed to evaluate the impact of sequencing cabazitaxel and 177Lu-PSMA-617 on survival outcomes in patients with mCRPC. PATIENTS AND METHODS This is a retrospective, multicenter, cohort study which included patients with mCRPC who received sequential treatment with 177Lu-PSMA-617 and cabazitaxel between January 2015 and December 2023. Primary outcome was progression-free survival-2 (PFS-2) RESULTS: A total of 68 patients with mCRPC who received sequential 177Lu-PSMA-617 and cabazitaxel were included in the study. The primary outcome, progression-free survival-2 (PFS-2), was similar in patients treated with 177Lu-PSMA-617 first (LU-CA) and those receiving cabazitaxel (CA-LU) first (10.8 and 11.7 months, respectively; p = 0.422). The median overall survival (OS) was also similar in the LU-CA and CA-LU groups (16.6 and 19.9 months, respectively; p = 0.917). The objective response rate (ORR) for 177Lu-PSMA-617 was 23.1 % when used first and 16.1 % after cabazitaxel. ORR for cabazitaxel was 25.6 % and 31.3 % when used as the first agent and when used after 177Lu-PSMA-617, respectively. CONCLUSIONS In conclusion, treatment sequencing between cabazitaxel and 177Lu-PSMA-617 did not significantly affect survival outcomes in patients with mCRPC. These findings suggest that both drugs can be effectively integrated into the mCRPC treatment paradigm without concerns about the effect of sequencing. However, prospective data are needed to optimize sequencing strategies and explore their impact on specific patient subgroups for more personalized care.
Collapse
Affiliation(s)
- Hatice Bolek
- Ankara University School of Medicine, Department of Medical Oncology, Ankara, Turkey; Ankara University Cancer Institute, Ankara, Turkey
| | - Satı Coskun Yazgan
- Ankara University School of Medicine, Department of Medical Oncology, Ankara, Turkey; Ankara University Cancer Institute, Ankara, Turkey
| | - Furkan Ceylan
- Bilkent City Hospital, Department of Medical Oncology, Ankara, Turkey
| | | | - Cagatay Arslan
- Izmir University of Economics, Medical Point Hospital, Izmir, Turkey
| | - Tülay Kuş
- Gaziantep University School of Medicine, Department of Medical Oncology, Gaziantep, Turkey
| | - Deniz Tural
- Koc University Medical Faculty, Department of Medical Oncology, Istanbul, Turkey
| | | | - Nuriye Ozlem Kucuk
- Ankara University School of Medicine, Department of Nuclear Medicine, Ankara, Turkey
| | | | - Elena Castro
- 12 de Octubre University Hospital, Department of Medical Oncology, Madrid, Spain
| | - Emre Yekedüz
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yüksel Ürün
- Ankara University School of Medicine, Department of Medical Oncology, Ankara, Turkey; Ankara University Cancer Institute, Ankara, Turkey.
| |
Collapse
|
43
|
Wu G, Chen C, Chang J, Fazlollahi F, Makary MS. Expanding the Scope of Interventional Oncology: Locoregional Therapies in Extrahepatic Malignancies. Cancers (Basel) 2025; 17:726. [PMID: 40075574 PMCID: PMC11899649 DOI: 10.3390/cancers17050726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Locoregional therapies (LRTs), including transarterial embolization (TAE), transarterial chemoembolization (TACE), and transarterial radioembolization (TARE), have become integral in the management of hepatocellular carcinoma (HCC) in recent decades and continue to shape evolving treatment strategies. While their role in liver tumor management is well established, their potential for treating extrahepatic malignancies is gaining increasing attention. Notably, growing research has highlighted the promising applications of TAE, TACE, and TARE in extrahepatic cancers such as glioblastoma (GBM), soft tissue sarcomas (STSs), prostate cancer (PCa), pancreatic cancer, and renal cell carcinoma (RCC). This review aims to explore these novel applications, providing a comprehensive summary of the current literature, examining clinical outcomes, and discussing future directions for integrating these techniques into broader oncologic treatment strategies. METHODS A systematic literature review was conducted focusing on LRTs such as TAE, TACE, and TARE in extrahepatic malignancies. Studies published between May 1998 and December 2024 were included, emphasizing outcomes in GBM, STS, PCa, pancreatic cancer, and RCC. Data extraction prioritized clinical outcomes, safety profiles, and procedural efficacy. RESULTS LRTs demonstrated significant potential in managing extrahepatic malignancies, with TAE, TACE, and TARE showing promising results in palliative management and tumor control. Across studies, these therapies exhibited varying degrees of success in improving progression-free survival and overall survival, with minimal systemic toxicity. CONCLUSIONS The expanding application of LRTs in extrahepatic malignancies highlights their transformative potential in interventional oncology. By offering targeted, minimally invasive treatment options, these modalities bridge critical gaps in managing tumors refractory to conventional therapies. Future research should focus on standardizing protocols, optimizing patient selection, and exploring combination therapies to maximize their clinical efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Mina S. Makary
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (G.W.); (C.C.); (J.C.)
| |
Collapse
|
44
|
Khorasanchi A, Hong F, Yang Y, Singer EA, Wang P, Li M, Zheng L, Monk P, Mortazavi A, Meng L. Overcoming drug resistance in castrate-resistant prostate cancer: current mechanisms and emerging therapeutic approaches. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:9. [PMID: 40051495 PMCID: PMC11883235 DOI: 10.20517/cdr.2024.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 03/09/2025]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is driven by a complex network of resistance mechanisms against standard-of-care therapies, resulting in poor long-term outcomes. This review offers a uniquely comprehensive and integrative perspective on these resistance pathways, systematically examining both androgen receptor (AR)-dependent factors (including AR overexpression, point mutations, glucocorticoid receptor signaling, splice variants, post-translational modifications, altered coregulators, and intratumoral hormone biosynthesis) and AR-independent pathways (such as neuroendocrine differentiation, lineage plasticity, and alternative growth factor signaling). We also highlight resistance mechanisms influencing immunotherapy, chemotherapy, radiopharmaceutical therapy and targeted therapy. By synthesizing emerging insights across these domains, this review not only clarifies the underlying biology of mCRPC resistance but also identifies key leverage points for more effective interventions. Building on this foundation, we propose a forward-looking framework for overcoming mCRPC drug resistance, emphasizing the importance of biomarker-guided patient selection, combination strategies that simultaneously target multiple resistance mechanisms, and novel therapies under investigation. These recommendations are intended to guide future clinical trial designs and research priorities that move beyond incremental improvements. Ultimately, this comprehensive synthesis aims to serve as a resource for clinicians and researchers to accelerate the development of durable, precision-based treatment strategies in mCRPC.
Collapse
Affiliation(s)
- Adam Khorasanchi
- Division of Hospital Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Feng Hong
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yuanquan Yang
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Eric A. Singer
- Division of Urologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Peng Wang
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Mingjia Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Linghua Zheng
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Paul Monk
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
45
|
Bondia-Bescós S, Martín-Marcuartu JJ, Robles-Barba JJ, Cortés-Romera M. Atypical prostate cancer recurrence in the vas deferens detected by [ 18F]F-piflufolastat PET/CT imaging. Actas Urol Esp 2025:501690. [PMID: 39938645 DOI: 10.1016/j.acuroe.2025.501690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/14/2025]
Affiliation(s)
- S Bondia-Bescós
- Nuclear Medicine-PET (ICS-IDI) Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - J J Martín-Marcuartu
- Nuclear Medicine-PET (ICS-IDI) Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - J J Robles-Barba
- Nuclear Medicine-PET (ICS-IDI) Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Cortés-Romera
- Nuclear Medicine-PET (ICS-IDI) Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
46
|
Madsen C, Fuglø D, Pedersen M, Broholm R, Østergren PB, Bisbjerg R, Kongsted P, Nielsen K, Haarmark C, Zacho H. Prospective Head-to-Head Comparison of 18F-PSMA PET/CT and 18F-NaF PET/CT for Assessing Bone Metastases in 160 Patients with Newly Diagnosed High-Risk Prostate Cancer. J Nucl Med 2025; 66:223-229. [PMID: 39778967 PMCID: PMC11800739 DOI: 10.2967/jnumed.124.268275] [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: 08/21/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Prostate-specific membrane antigen (PSMA) PET/CT is increasingly used for primary staging in prostate cancer (PC), mainly because of its improved accuracy in detecting lymph node metastases compared with conventional imaging. However, the diagnostic benefit of PSMA PET/CT for detecting bone metastases is less well established. This study compares the diagnostic accuracy of 18F-PSMA PET/CT and 18F-NaF PET/CT for detecting bone metastases in patients newly diagnosed with PC. Methods: This prospective study included patients with histologically confirmed high-risk PC. All participants were referred from the department of urology to 18F-NaF PET/CT and underwent 18F-PSMA PET/CT within 3 weeks. Images were reviewed by 2 nuclear medicine physicians unaware of the results of the other imaging modality. Presence or absence of bone metastases and number of metastatic lesions were recorded. A reference standard was established at the patient level based on agreement between the 2 imaging modalities. In cases of concordance, both modalities were deemed correct. In cases of discordance, additional follow-up scans were performed. Diagnostic performance metrics, including sensitivity, specificity, and accuracy, were calculated. Results: In total, 160 participants were included. Sensitivity, specificity, and accuracy for detecting bone metastases at the patient level were 0.98, 0.99, and 0.99, respectively, for 18F-PSMA PET/CT, and 0.91, 1.00, and 0.97, respectively, for 18F-NaF PET/CT. No significant differences were found. The concordance rate of bone metastases between 18F-NaF and 18F-PSMA PET/CT at the patient level was observed in 154 patients (96.3%). 18F-PSMA PET/CT tended to identify more bone metastases per patient than 18F-NaF PET/CT. Conclusion: Both 18F-NaF and 18F-PSMA PET/CT exhibit high diagnostic accuracy for detecting bone metastases in newly diagnosed high-risk PC patients. 18F-PSMA PET/CT may detect additional metastatic lesions compared with 18F-NaF PET/CT. Subsequent 18F-NaF PET/CT may be redundant if no bone metastases are found on 18F-PSMA PET/CT.
Collapse
Affiliation(s)
- Claus Madsen
- Department of Nuclear Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark;
| | - Dan Fuglø
- Department of Nuclear Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - Maria Pedersen
- Department of Nuclear Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - Rikke Broholm
- Department of Nuclear Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - Peter B Østergren
- Department of Urology, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Rasmus Bisbjerg
- Department of Urology, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - Per Kongsted
- Department of Oncology, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - Kayalvili Nielsen
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
- Department of Radiology, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark; and
| | - Christian Haarmark
- Department of Nuclear Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Helle Zacho
- Department of Nuclear Medicine, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
47
|
Naqvi SAA, Riaz IB. The Promise and Challenges of Genomic Classifiers in Localized Prostate Cancer. Ann Intern Med 2025; 178:290-291. [PMID: 39832371 DOI: 10.7326/annals-24-03630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Affiliation(s)
| | - Irbaz Bin Riaz
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| |
Collapse
|
48
|
Gillessen S, Turco F, Davis ID, Efstathiou JA, Fizazi K, James ND, Shore N, Small E, Smith M, Sweeney CJ, Tombal B, Zilli T, Agarwal N, Antonarakis ES, Aparicio A, Armstrong AJ, Bastos DA, Attard G, Axcrona K, Ayadi M, Beltran H, Bjartell A, Blanchard P, Bourlon MT, Briganti A, Bulbul M, Buttigliero C, Caffo O, Castellano D, Castro E, Cheng HH, Chi KN, Clarke CS, Clarke N, de Bono JS, De Santis M, Duran I, Efstathiou E, Ekeke ON, El Nahas TIH, Emmett L, Fanti S, Fatiregun OA, Feng FY, Fong PCC, Fonteyne V, Fossati N, George DJ, Gleave ME, Gravis G, Halabi S, Heinrich D, Herrmann K, Hofman MS, Hope TA, Horvath LG, Hussain MHA, Jereczek-Fossa BA, Jones RJ, Joshua AM, Kanesvaran R, Keizman D, Khauli RB, Kramer G, Loeb S, Mahal BA, Maluf FC, Mateo J, Matheson D, Matikainen MP, McDermott R, McKay RR, Mehra N, Merseburger AS, Morgans AK, Morris MJ, Mrabti H, Mukherji D, Murphy DG, Murthy V, Mutambirwa SBA, Nguyen PL, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Parker C, Poon DMC, Pritchard CC, Rabah DM, Rathkopf D, Reiter RE, Renard-Penna R, Ryan CJ, Saad F, Sade JP, Sandhu S, Sartor OA, Schaeffer E, Scher HI, et alGillessen S, Turco F, Davis ID, Efstathiou JA, Fizazi K, James ND, Shore N, Small E, Smith M, Sweeney CJ, Tombal B, Zilli T, Agarwal N, Antonarakis ES, Aparicio A, Armstrong AJ, Bastos DA, Attard G, Axcrona K, Ayadi M, Beltran H, Bjartell A, Blanchard P, Bourlon MT, Briganti A, Bulbul M, Buttigliero C, Caffo O, Castellano D, Castro E, Cheng HH, Chi KN, Clarke CS, Clarke N, de Bono JS, De Santis M, Duran I, Efstathiou E, Ekeke ON, El Nahas TIH, Emmett L, Fanti S, Fatiregun OA, Feng FY, Fong PCC, Fonteyne V, Fossati N, George DJ, Gleave ME, Gravis G, Halabi S, Heinrich D, Herrmann K, Hofman MS, Hope TA, Horvath LG, Hussain MHA, Jereczek-Fossa BA, Jones RJ, Joshua AM, Kanesvaran R, Keizman D, Khauli RB, Kramer G, Loeb S, Mahal BA, Maluf FC, Mateo J, Matheson D, Matikainen MP, McDermott R, McKay RR, Mehra N, Merseburger AS, Morgans AK, Morris MJ, Mrabti H, Mukherji D, Murphy DG, Murthy V, Mutambirwa SBA, Nguyen PL, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Parker C, Poon DMC, Pritchard CC, Rabah DM, Rathkopf D, Reiter RE, Renard-Penna R, Ryan CJ, Saad F, Sade JP, Sandhu S, Sartor OA, Schaeffer E, Scher HI, Sharifi N, Skoneczna IA, Soule HR, Spratt DE, Srinivas S, Sternberg CN, Suzuki H, Taplin ME, Thellenberg-Karlsson C, Tilki D, Türkeri LN, Uemura H, Ürün Y, Vale CL, Vapiwala N, Walz J, Yamoah K, Ye D, Yu EY, Zapatero A, Omlin A. Management of Patients with Advanced Prostate Cancer. Report from the 2024 Advanced Prostate Cancer Consensus Conference (APCCC). Eur Urol 2025; 87:157-216. [PMID: 39394013 DOI: 10.1016/j.eururo.2024.09.017] [Show More Authors] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Innovations have improved outcomes in advanced prostate cancer (PC). Nonetheless, we continue to lack high-level evidence on a variety of topics that greatly impact daily practice. The 2024 Advanced Prostate Cancer Consensus Conference (APCCC) surveyed experts on key questions in clinical management in order to supplement evidence-based guidelines. Here we present voting results for questions from APCCC 2024. METHODS Before the conference, a panel of 120 international PC experts used a modified Delphi process to develop 183 multiple-choice consensus questions on eight different topics. Before the conference, these questions were administered via a web-based survey to the voting panel members ("panellists"). KEY FINDINGS AND LIMITATIONS Consensus was a priori defined as ≥75% agreement, with strong consensus defined as ≥90% agreement. The voting results show varying degrees of consensus, as discussed in this article and detailed in the Supplementary material. These findings do not include a formal literature review or meta-analysis. CONCLUSIONS AND CLINICAL IMPLICATIONS The voting results can help physicians and patients navigate controversial areas of clinical management for which high-level evidence is scant or conflicting. The findings can also help funders and policymakers in prioritising areas for future research. Diagnostic and treatment decisions should always be individualised on the basis of patient and cancer characteristics, and should incorporate current and emerging clinical evidence, guidelines, and logistic and economic factors. Enrolment in clinical trials is always strongly encouraged. Importantly, APCCC 2024 once again identified important gaps (areas of nonconsensus) that merit evaluation in specifically designed trials.
Collapse
Affiliation(s)
- Silke Gillessen
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland.
| | - Fabio Turco
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Ian D Davis
- Monash University, Melbourne, Australia; Eastern Health, Melbourne, Australia
| | | | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | | | - Neal Shore
- Carolina Urologic Research Center and GenesisCare, Myrtle Beach, SC, USA
| | - Eric Small
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, CA, USA
| | - Matthew Smith
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Christopher J Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia
| | - Bertrand Tombal
- Division of Urology, Clinique Universitaire St. Luc, Brussels, Belgium
| | - Thomas Zilli
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Armstrong
- Center for Prostate and Urologic Cancer, Duke Cancer Institute, Duke University, Durham, NC, USA
| | | | | | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | - Mouna Ayadi
- Salah Azaiz Institute, Medical School of Tunis, Tunis, Tunisia
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Anders Bjartell
- Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Pierre Blanchard
- Department of Radiation Oncology, Oncostat U1018 INSERM, Université Paris-Saclay, Gustave-Roussy, Villejuif, France
| | - Maria T Bourlon
- Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Muhammad Bulbul
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Consuelo Buttigliero
- Department of Oncology, San Luigi Hospital, University of Turin, Orbassano, Italy
| | - Orazio Caffo
- Medical Oncology Department, Santa Chiara Hospital, APSS, Trento, Italy
| | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Heather H Cheng
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Kim N Chi
- BC Cancer and University of British Columbia, Vancouver, Canada
| | - Caroline S Clarke
- Research Department of Primary Care and Population Health, University College London, London, UK
| | - Noel Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - Johann S de Bono
- Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Ignacio Duran
- Medical Oncology Department, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Spain
| | | | - Onyeanunam N Ekeke
- Urology Division, University of Port Harcourt Teaching Hospital, Port Harcourt, Nigeria
| | | | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia; Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Stefano Fanti
- Department of Nuclear Medicine, IRCCS AOU Bologna, Bologna, Italy
| | | | - Felix Y Feng
- University of California-San Francisco, San Francisco, CA, USA
| | - Peter C C Fong
- Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | | | - Nicola Fossati
- Department of Surgery (Urology Service), Ente Ospedaliero Cantonale, Università della Svizzera Italiana Lugano, Switzerland
| | - Daniel J George
- Departments of Medicine and Surgery, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Martin E Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Daniel Heinrich
- Department of Oncology and Radiotherapy, Innlandet Hospital Trust, Gjøvik, Norway
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium, University Hospital Essen, Essen, Germany
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California-San Francisco, San Francisco, CA, USA
| | - Lisa G Horvath
- Chris O'Brien Lifehouse, University of Sydney, Sydney, Australia
| | - Maha H A Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy; Department of Radiation Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Robert J Jones
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Anthony M Joshua
- Department of Medical Oncology, Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, Australia
| | | | - Daniel Keizman
- Genitourinary Unit, Division of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Raja B Khauli
- Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon; Division of Urology, Carle-Illinois College of Medicine, Urbana, IL, USA
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Stacy Loeb
- Department of Urology and Population Health, New York University Langone Health, New York, NY, USA; Department of Surgery/Urology, Manhattan Veterans Affairs, New York, NY, USA
| | - Brandon A Mahal
- Department of Radiation Oncology, University of Miami Sylvester Cancer Center, Miami, FL, USA
| | - Fernando C Maluf
- Beneficiência Portuguesa de São Paulo, São Paulo, Brazil; Departamento de Oncologia, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Joaquin Mateo
- Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - David Matheson
- Faculty of Education Health and Wellbeing, University of Wolverhampton, Walsall, UK
| | - Mika P Matikainen
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Ray McDermott
- Department of Medical Oncology, St. Vincent's University Hospital and Cancer Trials, Dublin, Ireland
| | - Rana R McKay
- University of California-San Diego, Palo Alto, CA, USA
| | - Niven Mehra
- Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Alicia K Morgans
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hind Mrabti
- Institut National d'Oncologie, Mohamed V University, Rabat, Morocco
| | - Deborah Mukherji
- Clemenceau Medical Center, Dubai, United Arab Emirates; Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Vedang Murthy
- Radiation Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Shingai B A Mutambirwa
- Department of Urology, Sefako Makgatho Health Science University, Dr. George Mukhari Academic Hospital, Medunsa, South Africa
| | - Paul L Nguyen
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - William K Oh
- Division of Hematology and Medical Oncology, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, Antwerp, Belgium; Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Joe M O'Sullivan
- Patrick G. Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| | - Chris Parker
- Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Darren M C Poon
- Hong Kong Sanatorium and Hospital, Chinese University of Hong Kong, Hong Kong, China
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Danny M Rabah
- Cancer Research Chair and Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Urology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dana Rathkopf
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Raphaele Renard-Penna
- Department of Imagery, GRC 5 Predictive Onco-Uro, Pitie-Salpetriere Hospital, AP-HP, Sorbonne University, Paris, France
| | - Charles J Ryan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Fred Saad
- Centre Hospitalier de Université de Montréal, Montreal, Canada
| | | | - Shahneen Sandhu
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Oliver A Sartor
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| | - Edward Schaeffer
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nima Sharifi
- Desai Sethi Urology Institute and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Iwona A Skoneczna
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Sandy Srinivas
- Division of Medical Oncology, Stanford University Medical Center, Stanford, CA, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Division of Hematology and Oncology, Meyer Cancer Center, New York Presbyterian Hospital, New York, NY, USA
| | - Hiroyoshi Suzuki
- Department of Urology, Toho University Sakura Medical Center, Sakura, Japan
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Derya Tilki
- Martini-Klinik Prostate Cancer Center and Department of Urology, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Levent N Türkeri
- Department of Urology, M.A. Aydınlar Acıbadem University, Altunizade Hospital, Istanbul, Turkey
| | - Hiroji Uemura
- Yokohama City University Medical Center, Yokohama, Japan
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University School of Medicine, Ankara, Turkey
| | - Claire L Vale
- MRC Clinical Trials Unit, University College London, London, UK
| | - Neha Vapiwala
- Department of Radiation Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jochen Walz
- Institut Paoli-Calmettes Cancer Center, Marseille, France
| | - Kosj Yamoah
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Evan Y Yu
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA USA
| | - Almudena Zapatero
- University Hospital La Princesa, Health Research Institute, Madrid, Spain
| | - Aurelius Omlin
- Onkozentrum Zurich, University of Zurich and Tumorzentrum Hirslanden Zurich, Zurich, Switzerland
| |
Collapse
|
49
|
Abramczyk E, Nisar MU, Nguyen JK, Austin N, Ward RD, Weight C, Purysko AS. The Role of Prostate-Specific Membrane Antigen-Radioligand and Magnetic Resonance Imaging in Patients with Prostate Cancer Biochemical Recurrence. Semin Ultrasound CT MR 2025; 46:71-82. [PMID: 39580035 DOI: 10.1053/j.sult.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
A significant proportion of men with prostate cancer will experience biochemical recurrence (BCR), which is characterized by an elevation in prostate-specific antigen (PSA) levels after receiving treatment with curative intent. Imaging plays an important role in the management of patients with BCR. It can help identify sites of recurrence to determine the most appropriate management strategies, ranging from salvage treatment for local recurrences to systemic treatments for those with advanced, distant disease. PET/CT with prostate-specific membrane antigen (PSMA)-radioligands is the most sensitive method for the detection of prostate cancer recurrence, with significantly higher cancer detection rates compared to conventional imaging techniques such as bone scan and computed tomography, even at lower PSA levels. Nevertheless, interpretation of PSMA PET/CT images can be challenging, particularly for the evaluation of local recurrence due to urinary activity that can mimic or mask the presence of cancer. Furthermore, some prostate cancers may not express PSMA and have false negative results. Multiparametric prostate MRI is an excellent method for the evaluation of local recurrence and can overcome some of the limitations of PSMA PET/CT. In this review, we discuss the role of imaging in managing patients with prostate cancer BCR and describe the potential benefits of MRI in the PSMA-radioligand imaging era, emphasizing the assessment of local recurrence.
Collapse
Affiliation(s)
- Emily Abramczyk
- Department of Radiology, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
| | | | - Jane K Nguyen
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH
| | - Nicholas Austin
- Nuclear Medicine Department, Cleveland Clinic, Cleveland, OH; Abdominal Imaging Section, Cleveland Clinic, Cleveland, OH
| | - Ryan D Ward
- Abdominal Imaging Section, Cleveland Clinic, Cleveland, OH
| | - Christopher Weight
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH
| | - Andrei S Purysko
- Department of Radiology, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH; Nuclear Medicine Department, Cleveland Clinic, Cleveland, OH; Abdominal Imaging Section, Cleveland Clinic, Cleveland, OH; Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
50
|
Fountzilas E, Pearce T, Baysal MA, Chakraborty A, Tsimberidou AM. Convergence of evolving artificial intelligence and machine learning techniques in precision oncology. NPJ Digit Med 2025; 8:75. [PMID: 39890986 PMCID: PMC11785769 DOI: 10.1038/s41746-025-01471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025] Open
Abstract
The confluence of new technologies with artificial intelligence (AI) and machine learning (ML) analytical techniques is rapidly advancing the field of precision oncology, promising to improve diagnostic approaches and therapeutic strategies for patients with cancer. By analyzing multi-dimensional, multiomic, spatial pathology, and radiomic data, these technologies enable a deeper understanding of the intricate molecular pathways, aiding in the identification of critical nodes within the tumor's biology to optimize treatment selection. The applications of AI/ML in precision oncology are extensive and include the generation of synthetic data, e.g., digital twins, in order to provide the necessary information to design or expedite the conduct of clinical trials. Currently, many operational and technical challenges exist related to data technology, engineering, and storage; algorithm development and structures; quality and quantity of the data and the analytical pipeline; data sharing and generalizability; and the incorporation of these technologies into the current clinical workflow and reimbursement models.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, St Luke's Clinic, Panorama, Thessaloniki, Greece
| | | | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA.
| |
Collapse
|