1
|
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
|
2
|
Yan L, Su P, Sun X. Role of multi‑omics in advancing the understanding and treatment of prostate cancer (Review). Mol Med Rep 2025; 31:130. [PMID: 40116118 PMCID: PMC11938414 DOI: 10.3892/mmr.2025.13495] [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: 09/30/2024] [Accepted: 01/27/2025] [Indexed: 03/23/2025] Open
Abstract
The application of multi‑omics methodologies, encompassing genomics, transcriptomics, proteomics, metabolomics and integrative genomics, has markedly enhanced the understanding of prostate cancer (PCa). These methods have facilitated the identification of molecular pathways and biomarkers crucial for the early detection, prognostic evaluation and personalized treatment of PCa. Studies using multi‑omics technologies have elucidated how alterations in gene expression and protein interactions contribute to PCa progression and treatment resistance. Furthermore, the integration of multi‑omics data has been used in the identification of novel therapeutic targets and the development of innovative treatment modalities, such as precision medicine. The evolving landscape of multi‑omics research holds promise for not only deepening the understanding of PCa biology but also for fostering the development of more effective and tailored therapeutic interventions, ultimately improving patient outcomes. The present review aims to synthesize current findings from multi‑omics studies associated with PCa and to assess their implications for the improvement of patient management and therapeutic outcomes. The insights provided may guide future research directions and clinical practices in the fight against PCa.
Collapse
Affiliation(s)
- Li Yan
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Pengxiao Su
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaoke Sun
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
3
|
Bertozzi L, Zhang E, Behtaj M, Gordon O, Whalen MJ. Molecular profiling of basal cell carcinoma of the prostate: A case report and literature review. Urol Case Rep 2025; 60:102993. [PMID: 40124187 PMCID: PMC11925511 DOI: 10.1016/j.eucr.2025.102993] [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: 01/25/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025] Open
Abstract
Prostate basal cell carcinoma (BCC) is a rare pathologic variant with a poorly understood molecular profile. Here, we describe a case of prostate BCC and compare its genetic alterations to cases in the literature. After presenting with hematuria, our patient underwent definitive radical prostatectomy for his localized biopsy-proven BCC. Somatic and germline testing revealed mutations in PIK3R1, KMT2D, and NOTCH1, and MUTYH, NBN, and MSH3, respectively. Upon literature review, we found that prostate BCC mutations disrupt cell growth, epigenetic regulation, and cell fate determination. With no consensus guidelines available, experimental targeted therapies have shown promise for prostate BCC management.
Collapse
Affiliation(s)
- Luca Bertozzi
- The George Washington University School of Medicine and Health Sciences, 2300 I St NW, Washington, DC, 20052, USA
| | - Eileen Zhang
- University of Maryland, 7901 Regents Drive, College Park, MD, 20742, USA
| | - Mohadese Behtaj
- Department of Pathology, The George Washington Medical Faculty Associates, 2300 M Street NW, Suite 715, Washington, DC, 20037, USA
| | - Olivia Gordon
- Department of Urology, The George Washington Medical Faculty Associates, 2150 Pennsylvania Ave NW, Suite 3-417, Washington, DC, 20037, USA
| | - Michael J. Whalen
- Department of Urology, The George Washington Medical Faculty Associates, 2150 Pennsylvania Ave NW, Suite 3-417, Washington, DC, 20037, USA
| |
Collapse
|
4
|
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
|
5
|
Segalés L, Bellmunt J, Perera-Bel J, Vargas-Parra G, Juanpere N, López D, Rodriguez-Vida A, Colomo L, Cecchini L, Lloreta-Trull J, Yélamos J, Fumadó L, Hernández-Llodrà S. Prognostic value of PARP1 and PARP2 copy number alterations in prostate cancer. J Transl Med 2025:104171. [PMID: 40210166 DOI: 10.1016/j.labinv.2025.104171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
PARP1/2 have overlapping yet non-redundant biological functions in DNA repair and AR-transcriptional regulation. Studies on PARP alterations in human tumors have yielded conflicting results. In prostate cancer (PCa), PARP1/2 protein overexpression has been related to androgen deprivation therapy (ADT) resistance, biochemical recurrence, and progression to metastases. PARP inhibitors (PARPi) have been approved for treating metastatic castration-resistant PCa with homologous recombination repair (HRR) gene mutations. However, the significance of PARP1/2 genomic alterations is not fully studied. We aimed to analyze PARP1/2 alterations in PCa, to assess their value as prognostic markers, and to explore their relevance for potential therapeutic stratification. PARP1/2 copy number status was evaluated in 121 PCa primary tumors through real time PCR. In 29 of them, a regional pelvic lymph node (RLN) involvement was also analyzed. BRCA1/2 somatic mutations were analyzed in 24 PCa. Relationship with clinicopathological features, progression to metastases, and PSA recurrence was assessed. PARP1 loss and PARP2 gain were detected in 34.7% and 32.2% of primary tumors, respectively, with a high frequency of co-occurrence (p<0.001). Both alterations were statistically associated with locally-advanced disease at diagnosis (p=0.036; p=0.006), metastatic dissemination (p=0.014; p=0.003), and other aggressive clinicopathological characteristics (as presence of Gleason pattern 5, high-grade, and high-stage). Cases with exclusive PARP2 gain had the shortest time to PSA recurrence, while double wt patients displayed the best outcome (p=0.007). In 29 paired primary tumors and RLN involvement, PARP1 loss showed strong concordance (p=0.001), whereas PARP2 gain did not (p=0.411). In conclusion, loss of PARP1 and gain of PARP2 show strong co-occurrence, and are associated with clinicopathological characteristics of aggressiveness. PARP2 alterations appear to have a particularly significant impact on disease prognosis. Furthermore, these data suggest that the analysis of PARP1/2 copy number status could be useful to predict PCa outcome. Its role on therapy warrants further evaluation.
Collapse
Affiliation(s)
- Laura Segalés
- Departament of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain
| | - Joaquim Bellmunt
- Hospital del Mar Research Institute, Barcelona, Spain; Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | - Nuria Juanpere
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - David López
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Alejo Rodriguez-Vida
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital del Mar, Barcelona, Spain
| | - Lluís Colomo
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Lluís Cecchini
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Urology, Hospital del Mar, Barcelona, Spain
| | - Josep Lloreta-Trull
- Departament of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - José Yélamos
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Lluís Fumadó
- Hospital del Mar Research Institute, Barcelona, Spain; Department of Urology, Hospital del Mar, Barcelona, Spain
| | - Silvia Hernández-Llodrà
- Departament of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
6
|
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
|
7
|
Rosar F, Khreish F, Nagel LS, Blickle A, Burgard C, Petto S, Bastian MB, Speicher T, Bartholomä M, Maus S, Schaefer-Schuler A, Ezziddin S. 225 Ac-PSMA-617 Augmentation After Insufficient Response Under 177 Lu-PSMA-617 Radioligand Therapy in mCRPC: Evaluation of Outcome and Safety From a Prospective Registry (REALITY Study). Clin Nucl Med 2025; 50:e202-e206. [PMID: 39787407 DOI: 10.1097/rlu.0000000000005640] [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: 01/12/2025]
Abstract
BACKGROUND Even though the introduction of 177 Lu-PSMA-617 RLT represents a major milestone in the treatment of mCRPC, there are still patients who do not respond adequately to this therapy and for whom there are only limited options left. Augmenting 177 Lu-PSMA-617 RLT with the alpha-emitter 225 Ac-PSMA-617 may present an escalating treatment option to increase efficacy. In this study, we aim to evaluate outcome and safety of 225 Ac-PSMA-617 augmentation to 177 Lu-PSMA-617 RLT in patients who present insufficient response to monotherapy with 177 Lu-PSMA-617 RLT. PATIENTS AND METHODS The study included n = 51 mCRPC patients enrolled in a prospective registry receiving 177 Lu-PSMA-617 monotherapy and showing insufficient response, followed by initiation of 225 Ac-PSMA-617 augmentation, with adjusted activities depending on individual patient characteristics. Biochemical response, progression-free survival, and overall survival were assessed. Adverse events were evaluated according to the Common Terminology Criteria for Adverse Events (CTCAE v.5.0). RESULTS After initiation of 225 Ac-PSMA-617 augmentation to 177 Lu-PSMA-617 RLT, 24/51 patients (47.1%) exhibited partial remission, 16/51 (31.4%) stable disease, and 11/51 (21.6%) progressive disease. The median progression-free survival and overall survival rates were 6.3 months and 9.1 months, respectively. The majority of CTCAE gradings remained stable after initiating augmentation. Severe adverse events were rare, and no treatment termination due to side effects was recorded. CONCLUSIONS 225 Ac-PSMA-617 augmented 177 Lu-PSMA-617 radioligand therapy seems to be an effective escalating treatment option in patients after failure of conventional 177 Lu-PSMA-617 RLT and represents a promising approach balancing antitumor effect and tolerable side effects.
Collapse
Affiliation(s)
- Florian Rosar
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | | | - Lea Sophie Nagel
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Arne Blickle
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Caroline Burgard
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Sven Petto
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Moritz B Bastian
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Tilman Speicher
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Mark Bartholomä
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Stephan Maus
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Andrea Schaefer-Schuler
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| | - Samer Ezziddin
- From the Department of Nuclear Medicine, Saarland University-Medical Center, Homburg, Germany
| |
Collapse
|
8
|
Séguier D, Parent P, Duterque-Coquillaud M, Labreuche J, Fromont-Hankard G, Dariane C, Penel N, Villers A, Turpin A, Olivier J. Emergence of Neuroendocrine Tumors in Patients Treated with Androgen Receptor Pathway Inhibitors for Metastatic Prostate Cancer: A Systematic Review and Meta-analysis. Eur Urol Oncol 2025; 8:581-590. [PMID: 39824723 DOI: 10.1016/j.euo.2024.12.014] [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: 09/24/2024] [Revised: 11/28/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND AND OBJECTIVE It has been shown that androgen receptor pathway inhibitor (ARPIs) treatment for metastatic castration-resistant prostate cancer (mCRPC) improves overall survival rates, but ARPIs appear to be associated with a higher frequency of treatment-related neuroendocrine prostate cancer (t-NEPC). Our aim was to quantify the proportion of prostate adenocarcinoma cases that transition to t-NEPC following ARPI therapy. METHODS We conducted a comprehensive search of the literature on t-NEPC using databases including MEDLINE and Scopus. Eligible studies reported outcome data for NEPC in patients with prior mCRPC treated with an ARPI. To determine the pooled frequency of neuroendocrine transformation, the Freeman-Tukey variance-stabilizing arcsine transformation was applied to individual frequencies. KEY FINDINGS AND LIMITATIONS Among the 938 patients in eight eligible studies, t-NEPC diagnosis was confirmed in 171 patients, predominantly via pathology. Baseline biopsy verification to ensure the absence of NEPC was performed in most cases. The definition of t-NEPC varied among the studies. Five studies used a morphological definition based on histopathology, and three studies used NEPC biomarker detection on circulating tumor cells. A meta-analysis of aggregate data revealed an overall NEPC frequency following ARPI therapy of 16% (95% confidence interval 9-24%). CONCLUSION AND CLINICAL IMPLICATIONS ARPI-related NEPC represents a frequently underdiagnosed late complication of mCRPC. Given the absence of biomarkers for diagnosis, routine repeat biopsy at the mCRPC stage should be considered to diagnose t-NEPC transitions.
Collapse
Affiliation(s)
- Denis Séguier
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Urology, Hospital Claude Huriez, CHU Lille, Lille, France
| | - Pauline Parent
- Department of Medical Oncology, CHU Lille, Lille, France
| | - Martine Duterque-Coquillaud
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France
| | | | | | - Charles Dariane
- Service d'Urologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Université Paris Cité, Paris, France
| | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, University of Lille, Lille, France
| | - Arnauld Villers
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Urology, Hospital Claude Huriez, CHU Lille, Lille, France
| | - Anthony Turpin
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Medical Oncology, CHU Lille, Lille, France
| | - Jonathan Olivier
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Urology, Hospital Claude Huriez, CHU Lille, Lille, France.
| |
Collapse
|
9
|
Dai YH, Chen PH, Lee DJ, Andrade G, Vallis KA. A Meta-Analysis and Meta-Regression of the Efficacy, Toxicity, and Quality of Life Outcomes Following Prostate-Specific Membrane Antigen Radioligand Therapy Utilising Lutetium-177 and Actinium-225 in Metastatic Prostate Cancer. Eur Urol 2025; 87:398-408. [PMID: 39327114 DOI: 10.1016/j.eururo.2024.09.020] [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: 04/25/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND OBJECTIVE Management of metastatic prostate cancer (mPCa) presents significant challenges. In this systematic review, meta-analysis, and meta-regression, the efficacy, safety, and quality of life (QoL) outcomes of prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (PRLT) utilising lutetium-177 ([177Lu]Lu-PSMA) and actinium-225 ([225Ac]Ac-PSMA) were assessed. METHODS A detailed literature search across PubMed/Medline, EMBASE, Web of Science, Scopus, and Cochrane Library was conducted, culminating in the inclusion of 100 studies involving 8711 patients. Data on prostate-specific antigen (PSA) responses, toxicity profiles, and QoL and survival outcomes were analysed. Proportional meta-analyses and meta-regression analyses were performed. KEY FINDINGS AND LIMITATIONS The estimated proportion of patients with PSA decline ≥50% was 0.49 for [177Lu]Lu-PSMA and 0.60 for [225Ac]Ac-PSMA in mPCa, particularly metastatic castration-resistant prostate cancer. A meta-regression analysis indicated an association between the cumulative amount of administered activity and the proportion of PSA ≥50% decline. Positive PSA responses were observed alongside improved overall survival across both therapies. Our analyses also identified the key factors associated with PSA responses and survival outcomes, including baseline haemoglobin level, and the presence of visceral metastases. Although anaemia was commonly observed, with [177Lu]Lu-PSMA, severe toxicities were infrequent. Improved QoL was observed following [177Lu]Lu-PSMA therapy, whereas it remained stable following the second cycle of [225Ac]Ac-PSMA treatment. Heterogeneity across studies for PSA responses and toxicity profiles is a limitation. CONCLUSIONS AND CLINICAL IMPLICATIONS Our findings suggest an association between PRLT and reductions in PSA levels, as well as associations with enhanced survival outcomes in mPCa. Furthermore, our analysis shows a low incidence of severe toxicity associated with this treatment. These observations highlight the important role of PRLT in the management of mPCa.
Collapse
Affiliation(s)
- Yang-Hong Dai
- Department of Oncology, University of Oxford, Oxford, UK; Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Huang Chen
- Division of Haematology and Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ding-Jie Lee
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital Keelung Branch, National Defense Medical Center, Taiwan; Department of Biological Science and Technology, Institute of Bioinformatics and System Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Gerard Andrade
- Department of Clinical Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | |
Collapse
|
10
|
Castro E, Orji C, Ribbands A, Butcher J, Walley M, Li W, Ghate SR. Real-world treatment patterns and genetic testing in a metastatic castration-resistant prostate cancer setting in Europe. Future Oncol 2025; 21:1085-1099. [PMID: 40105456 DOI: 10.1080/14796694.2025.2470616] [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: 12/17/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
AIM This study described treatment patterns, reasons for treatment, and homologous recombination repair mutation (HRRm) testing patterns in a real-world metastatic castration-resistant prostate cancer (mCRPC) population in Europe. METHODS Data were drawn from the Adelphi Prostate Cancer Disease Specific Programme™, a cross-sectional survey of physicians and patients conducted in France, Germany, Italy, Spain, and the United Kingdom, November 2022-May 2023. Physicians provided clinical characteristics, treatment and HRRm testing patterns, and reasons for treatment for eight consecutive patients with mCRPC. Most analyses were descriptive; treatment reasons were compared using Fisher's Exact test. RESULTS Physicians provided data for 1,737 mCRPC patients. Most patients (73%) were androgen receptor pathway inhibitor (ARPi)-naïve at first-line (1 L) mCRPC. Here, at 1 L mCRPC, 60% of patients received ARPi and 24% chemotherapy. Of those who received ARPi prior to mCRPC (n = 291), 60% received chemotherapy at mCRPC and 21% ARPi. Overall, 37% were HRRm tested. Treatment patterns, sequencing, reasons, and HRRm testing varied by country, physician specialty, and practice setting. CONCLUSIONS Treatment patterns generally followed guidelines. ARPi prescriptions prior to mCRPC were numerically higher than previously reported, however HRRm testing rates were still low meaning mCRPC patients may miss out on more effective targeted treatments.
Collapse
Affiliation(s)
- Elena Castro
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Chinelo Orji
- Outcomes Research, Merck & Co. Inc., Rahway, NJ, USA
| | | | - Jake Butcher
- Oncology Team, Adelphi Real World, Bollington, UK
| | - Maria Walley
- Oncology Team, Adelphi Real World, Bollington, UK
| | - Weiyan Li
- Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD, USA
| | | |
Collapse
|
11
|
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] [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
|
12
|
Wang C, Han X, Kong S, Zhang S, Ning H, Wu F. Deciphering the mechanisms of PARP inhibitor resistance in prostate cancer: Implications for precision medicine. Biomed Pharmacother 2025; 185:117955. [PMID: 40086424 DOI: 10.1016/j.biopha.2025.117955] [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: 01/01/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Prostate cancer is a leading malignancy among men. While early-stage prostate cancer can be effectively managed, metastatic prostate cancer remains incurable, with a median survival of 3-5 years. The primary treatment for advanced prostate cancer is androgen deprivation therapy (ADT), but resistance to ADT often leads to castrationresistant prostate cancer (CRPC), presenting a significant therapeutic challenge. The advent of precision medicine has introduced promising new treatments, including PARP inhibitors (PARPi), which target defects in DNA repair mechanisms in cancer cells. PARPi have shown efficacy in treating advanced prostate cancer, especially in patients with metastatic CRPC (mCRPC) harboring homologous recombination (HR)-associated gene mutations. Despite these advancements, resistance to PARPi remains a critical issue. Here, we explored the primary mechanisms of PARPi resistance in prostate cancer. Key resistance mechanisms include homologous recombination recovery through reverse mutations in BRCA genes, BRCA promoter demethylation, and non-degradation of mutated BRCA proteins. The tumor microenvironment and overactivation of the base excision repair pathway also play significant roles in bypassing PARPi-induced synthetic lethality. In addition, we explored the clinical implications and therapeutic strategies to overcome resistance,emphasizing the need for precision medicine approaches. Our findings highlight the need for comprehensive strategies to improve PARPi sensitivity and effectiveness,ultimately aiming to extend patient survival and improve the quality of life for those with advanced prostate cancer. As our understanding of PARPi resistance evolves, more diverse and effective individualized treatment regimens will emerge.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China
| | - Xiaoran Han
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shaoqiu Kong
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shanhua Zhang
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Hao Ning
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| | - Fei Wu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| |
Collapse
|
13
|
García-Díaz HC, Larrosa-Garcia M, Gómez-Alonso J, Cruellas M, Felip E, Macarulla T, Farriols A, Carreras MJ. Off-label use of olaparib in uncommon tumor locations in patients with impaired homologous recombination genes. FARMACIA HOSPITALARIA 2025:S1130-6343(25)00017-0. [PMID: 40155245 DOI: 10.1016/j.farma.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/28/2025] [Accepted: 02/18/2025] [Indexed: 04/01/2025] Open
Abstract
OBJECTIVE To describe the effectiveness and safety of olaparib off-label indications in patients with impaired homologous recombination genes and solid tumors different than those authorized. METHODS A single-center, observational and retrospective study including patients treated with olaparib for off-label use. The main variables were patient characteristics, prior therapies, response to therapy, progression-free survival, overall survival and adverse events. RESULTS A total of 6 patients were included. All patients had metastases and received 3 or more lines of prior treatment. The primary tumor locations and mutations were partner and localizer of BRCA2 (PALB2) intrahepatic cholangiocarcinoma, ataxia telangiectasia mutated (ATM) non-small cell lung adenocarcinoma, somatic breast cancer gene (sBRCA2) colorectal cancer, germinal breast cancer gene 2 (gBRCA2) breast neuroendocrine tumor, gBRCA2 ampullary cancer and gBRCA2 pancreatic neuroendocrine tumor. At the end of the study, one patient was still receiving olaparib showing more than 25 months of sustained stable disease response. No novel toxicities were observed besides those included in the product information. CONCLUSIONS There is limited published evidence on the use of olaparib in patients harboring pathogenic variants other than breast cancer genes, like PALB2 and ATM and conditions different than those authorized such as digestive tract, neuroendocrine and lung tumors. Further research is to assess the efficacy of olaparib in these patients.
Collapse
Affiliation(s)
| | | | - Javier Gómez-Alonso
- Department of Pharmacy, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Mara Cruellas
- Medical Oncology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Teresa Macarulla
- Medical Oncology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Anna Farriols
- Department of Pharmacy, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Maria J Carreras
- Department of Pharmacy, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| |
Collapse
|
14
|
McHugh J, Bancroft E, Kote-Jarai Z, Eeles R. Prostate Cancer: genetics in practice now and in the future. Hered Cancer Clin Pract 2025; 23:11. [PMID: 40134022 PMCID: PMC11938723 DOI: 10.1186/s13053-025-00310-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/27/2025] Open
Abstract
Prostate Cancer (PrCa) is one of the most common cancers worldwide and causes a significant healthcare burden. Recent predictions estimate the incidence of new cases of PrCa will double from 1.4 million in 2020 to 2.9 million by 2040.The known risk factors for PrCa are increasing age, family history, ancestry and genetics. PrCa is one of the most heritable of the more common cancers. The heritability of PrCa is due to both rare moderate to high-risk monogenic variants and more common variants known as single nucleotide polymorphisms (SNPs) which can be used to calculate a polygenic risk score (PRS) for PrCa, while there is some of the genetic risk as yet unexplained. In recent years more PrCa risk-associated SNPs have been identified, increasing over time with the inclusion of more persons of diverse ancestry in studies. The identification of germline variants known to be associated with increased PrCa risk and disease aggressiveness has led to targeted treatments for certain pathogenic variant carriers.This is a mini review of how the genetics of PrCa can impact on screening and early detection of the disease and the treatment and management of the disease when diagnosed.
Collapse
Affiliation(s)
- Jana McHugh
- The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK.
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK.
| | - Elizabeth Bancroft
- The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Rosalind Eeles
- The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
15
|
Kazmi F, Shrestha N, Liu TFD, Foord T, Heesen P, Booth S, Dodwell D, Lord S, Yeoh KW, Blagden SP. Next-generation sequencing for guiding matched targeted therapies in people with relapsed or metastatic cancer. Cochrane Database Syst Rev 2025; 3:CD014872. [PMID: 40122129 PMCID: PMC11930395 DOI: 10.1002/14651858.cd014872.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
BACKGROUND Matched targeted therapies (MTT) given alone or in combination with systemic anti-cancer therapies have delivered proven survival benefit for many people with newly diagnosed cancer. However, there is little evidence of their effectiveness in the recurrent or late-stage setting. With this uncertainty, alongside the perception that late-stage cancers are too genetically heterogenous or too mutationally diverse to benefit from matched targeted therapies, next-generation sequencing (NGS) of tumours in people with refractory cancer remains a low priority. As a result, next-generation sequencing testing of recurrent or late-stage disease is discouraged. We lack evidence to support the utility of next generation sequencing in guiding matched targeted therapies in this setting. OBJECTIVES To evaluate the benefits and harms of matched targeted therapies in people with advanced cancers in randomised controlled trials. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, ClinicalTrials.gov, and the World Health Organisation International Clinical Trials Registry Platform (WHO-ICTRP) search portal up to 30th October 2024. We also screened reference lists of included studies and also the publications that cited these studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) that had enroled participants with advanced/refractory solid or haematological cancers who had progressed through at least one line of standard anti-cancer systemic therapy. To be eligible, all participants should have received matched targeted therapy based on next-generation sequencing carried out on their tumour (tumour tissue, blood or bone marrow). DATA COLLECTION AND ANALYSIS We systematically searched medical databases (e.g. MEDLINE, Embase) and trial registers for randomised controlled trials (RCTs). Outcomes of interest were progression-free survival (PFS), overall survival (OS), overall response rates (ORR), serious (grade 3 or 4) adverse events (AEs) and quality of life (QOL). We used a random-effects model to pool outcomes across studies and compared predefined subgroups using interaction tests. Grading of Recommendations Assessment, Development and Evaluation (GRADE) assessment of certainty was used to evaluate the quality of evidence. MAIN RESULTS We identified a total of 37 studies, out of which 35 studies (including 9819 participants) were included in the meta-analysis. All included studies compared a matched targeted therapy intervention to standard-of-care treatment, non-matched targeted therapies or no treatment (best supportive care): Matched targeted therapy versus standard-of-care treatment Matched targeted therapy (MTT) compared with standard systematic therapy probably reduces the risk of disease progression by 34% (hazard ratio (HR) = 0.66, 95% confidence interval (CI) 0.59 to 0.74; 14 studies, 3848 participants; moderate-certainty evidence). However, MTT might have little to no difference in risk of death (HR = 0.85, 95% CI 0.75 to 0.97; 14 studies, 3848 participants; low-certainty evidence) and may increase overall response rates (low-certainty evidence). There was no clear evidence of a difference in severe (grade 3/4) adverse events between matched targeted therapy and standard-of-care treatment (low-certainty evidence). There was limited evidence of a difference in quality of life between groups (very low-certainty of evidence). Matched targeted therapy in combination with standard-of-care treatment versus standard-of-care treatment alone Matched targeted therapy in combination with standard-of-care treatment compared with standard-of-care treatment alone probably reduces the risk of disease progression by 39% (HR = 0.61, 95% CI 0.53-0.70, 14 studies, 2,637 participants; moderate-certainty evidence) and risk of death by 21% (HR = 0.79, 95% CI 0.70 to 0.89; 11 studies, 2575 participants, moderate-certainty evidence). The combination of MTT and standard-of-care treatment may also increase overall response rates (low-certainty evidence). There was limited evidence of a difference in the incidence of severe adverse events (very low-certainty evidence) and quality of life between the groups (very low-certainty of evidence). Matched targeted therapy versus non-matched targeted therapy Matched targeted therapy compared with non-matched targeted therapy probably reduces the risk of disease progression by 24% (HR = 0.76, 95% CI 0.64 to 0.89; 3 studies, 1568 participants; moderate-certainty evidence) and may reduce the risk of death by 25% (HR = 0.75, 95% CI 0.65 to 0.86, 1307 participants; low-certainty evidence). There was little to no effect on overall response rates between MTT and non-MTT. There was no clear evidence of a difference in overall response rates (low-certainty evidence) and severe adverse events between MTT and non-MTT (low-certainty evidence). None of the studies comparing MTT and non-MTT reported quality of life. Matched targeted therapy versus best supportive care Matched targeted therapy compared with the best supportive care (BSC) i.e. no active treatment probably reduces the risk of disease progression by 63% (HR 0.37, 95% CI 0.28 to 0.50; 4 studies, 858 participants; moderate-certainty evidence). There was no clear evidence of a difference in overall survival between groups (HR = 0.88, 95% CI 0.73 to 1.06, 3 studies, 783 participants; low-certainty evidence). There was no clear evidence of a difference in overall response rates (very low-certainty of evidence) and incidence of severe adverse events (very low-certainty of evidence) between the groups. Quality of life was reported in a single study but did not provide composite scores. Risk of bias The overall risk of bias was judged low for eight studies, unclear for two studies, and the remaining 27 studies were high risk. AUTHORS' CONCLUSIONS Matched targeted therapies guided by next-generation sequencing in people with advanced cancer prolongs the time before cancer progresses compared to standard therapies. However, there is limited evidence to suggest that it prolongs overall survival, improves the quality of life or increases adverse events. Importantly, this review supports equitable access to next-generation sequencing technology for all people with advanced cancer and offers them the opportunity to access genotype-matched targeted therapies.
Collapse
Affiliation(s)
- Farasat Kazmi
- Department of Oncology, University of Oxford, Oxford, UK
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Nipun Shrestha
- Health Evidence Synthesis, Recommendations and Impact (HESRI), School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Tik Fung Dave Liu
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | | | | | - Stephen Booth
- Department of Haematology, Royal Berkshire Hospital, Reading, UK
| | - David Dodwell
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Oxford, UK
| | - Kheng-Wei Yeoh
- Radiation Oncology, National Cancer Centre, Singapore, Singapore
| | | |
Collapse
|
16
|
Hofstad M, Woods A, Parra K, Sychev ZE, Mazzagatti A, Huo X, Yu L, Gilbreath C, Chen WM, Davis AJ, Ly P, Drake JM, Kittler R. Dual inhibition of ATR and DNA-PKcs radiosensitizes ATM-mutant prostate cancer. Oncogene 2025:10.1038/s41388-025-03343-x. [PMID: 40119228 DOI: 10.1038/s41388-025-03343-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/31/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025]
Abstract
In advanced castration resistant prostate cancer (CRPC), mutations in the DNA damage response (DDR) gene ataxia telangiectasia mutated (ATM) are common. While poly(ADP-ribose) polymerase inhibitors are approved in this context, their clinical efficacy remains limited. Thus, there is a compelling need to identify alternative therapeutic avenues for ATM mutant prostate cancer patients. Here, we generated matched ATM-proficient and ATM-deficient CRPC lines to elucidate the impact of ATM loss on DDR in response to DNA damage via irradiation. Through unbiased phosphoproteomic screening, we unveiled that ATM-deficient CRPC lines maintain dependence on downstream ATM targets through activation of ATR and DNA-PKcs kinases. Dual inhibition of ATR and DNA-PKcs effectively inhibited downstream γH2AX foci formation in response to irradiation and radiosensitized ATM-deficient lines to a greater extent than either ATM-proficient controls or single drug treatment. Further, dual inhibition abrogated residual downstream ATM pathway signaling and impaired replication fork dynamics. To circumvent potential toxicity, we leveraged the RUVBL1/2 ATPase inhibitor Compound B, which leads to the degradation of both ATR and DNA-PKcs kinases. Compound B effectively radiosensitized ATM-deficient CRPC in vitro and in vivo, and impacted replication fork dynamics. Overall, dual targeting of both ATR and DNA-PKcs is necessary to block DDR in ATM-deficient CRPC, and Compound B could be utilized as a novel therapy in combination with irradiation in these patients.
Collapse
Affiliation(s)
- Mia Hofstad
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Andrea Woods
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Karla Parra
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Zoi E Sychev
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Alice Mazzagatti
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Xiaofang Huo
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lan Yu
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Collin Gilbreath
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Wei-Min Chen
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anthony J Davis
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Peter Ly
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Justin M Drake
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA.
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
17
|
Liu S, Fu S, Wu X, Wu S, Zhao Y, Wu X, Yan L, Lu J, Li L, Tao Y. TAK-901, a novel EPHA2 inhibitor as a therapeutic strategy against prostate cancer. Cell Signal 2025; 131:111750. [PMID: 40101850 DOI: 10.1016/j.cellsig.2025.111750] [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: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Prostate cancer is the most common cancer and remains a leading cause of cancer-related deaths among men worldwide. Androgen deprivation therapy continues to be the cornerstone of treatment for prostate cancer. However, the efficacy of this treatments is often limited, leading to the emergence of drug resistance and tumor recurrence. TAK-901, an inhibitor of Aurora kinase B, has been shown to inhibit tumor growth both in vitro and in vivo models. To date, the effect of TAK-901 on prostate cancer and the underlying mechanism remain unknown. In this study, we found that TAK-901 could inhibit proliferation, colony formation and migration, while also inducing apoptosis in prostate cancer cells. We further demonstrated that TAK-901 activates the CHK1 signaling pathway, leading to G2/M-phase arrest in these cells. Additionally, we identified EPHA2 as a novel therapeutic target of TAK-901. By mutating the binding sites between EPHA2 and TAK-901, we discovered that these mutations could reverse the anti-proliferative effects of TAK-901 in prostate cancer models. Our study is the first to reveal that TAK-901 induces apoptosis in prostate cancer cells and inhibits cell growth by targeting EPHA2. These findings provide valuable insights into the underlying mechanisms of TAK-901 and may develop its therapeutic applications in prostate cancer.
Collapse
Affiliation(s)
- Shanhui Liu
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Shengjun Fu
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Xuewu Wu
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Shan Wu
- Gansu Provincial Center for Disease Control and Prevention, Lanzhou 730000, Gansu, China
| | - Youli Zhao
- Department of Clinical Medical Laboratory, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Xinyue Wu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Liting Yan
- Central Laboratory, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi, China
| | - Jianzhong Lu
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China.
| | - Lanlan Li
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China.
| | - Yan Tao
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China.
| |
Collapse
|
18
|
Shahani A, Slika H, Elbeltagy A, Lee A, Peters C, Dotson T, Raj D, Tyler B. The epigenetic mechanisms involved in the treatment resistance of glioblastoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:12. [PMID: 40201311 PMCID: PMC11977385 DOI: 10.20517/cdr.2024.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 04/10/2025]
Abstract
Glioblastoma (GBM) is an aggressive malignant brain tumor with almost inevitable recurrence despite multimodal management with surgical resection and radio-chemotherapy. While several genetic, proteomic, cellular, and anatomic factors interplay to drive recurrence and promote treatment resistance, the epigenetic component remains among the most versatile and heterogeneous of these factors. Herein, the epigenetic landscape of GBM refers to a myriad of modifications and processes that can alter gene expression without altering the genetic code of cancer cells. These processes encompass DNA methylation, histone modification, chromatin remodeling, and non-coding RNA molecules, all of which have been found to be implicated in augmenting the tumor's aggressive behavior and driving its resistance to therapeutics. This review aims to delve into the underlying interactions that mediate this role for each of these epigenetic components. Further, it discusses the two-way relationship between epigenetic modifications and tumor heterogeneity and plasticity, which are crucial to effectively treat GBM. Finally, we build on the previous characterization of epigenetic modifications and interactions to explore specific targets that have been investigated for the development of promising therapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Betty Tyler
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
19
|
Zhao J, Tang B, Shen P, Zeng H, Wei Q. Empowering PARP inhibition through rational combination: Mechanisms of PARP inhibitors and combinations with a focus on the treatment of metastatic castration-resistant prostate cancer. Crit Rev Oncol Hematol 2025; 210:104698. [PMID: 40089046 DOI: 10.1016/j.critrevonc.2025.104698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have revolutionized the treatment of many cancers. Metastatic castration-resistant prostate cancer (mCRPC) is an area where PARP inhibitors are intensively studied; the efficacy with PARP inhibitor monotherapy in patients with homologous recombination repair mutations following novel hormonal therapy have prompted the investigation of combination therapy, with adding an androgen receptor pathway inhibitor (ARPI) being one focus of research. Data on PARP inhibitor monotherapy and combination therapy for mCRPC are accumulating, and it is important to navigate through the complex data to inform treatment decision. Here we review the mechanisms of action of PARP inhibitors, their pharmacological properties, the synergistic activity of PARP inhibitors plus other drug classes, and the clinical evidence on monotherapy and combination therapy in patients with mCRPC. We propose key considerations in the selection of agents and treatment sequence for mCRPC, such as efficacy, toxicity profiles, biomarkers, and interactions with concomitant medications.
Collapse
Affiliation(s)
- Jinge Zhao
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Tang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
Wang YW, Allen I, Funingana G, Tischkowitz M, Joko-Fru YW. Predictive biomarkers for the efficacy of PARP inhibitors in ovarian cancer: an updated systematic review. BJC REPORTS 2025; 3:14. [PMID: 40069561 PMCID: PMC11897386 DOI: 10.1038/s44276-025-00122-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/06/2024] [Accepted: 01/09/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND PARP inhibitors are effective in treating ovarian cancer, especially for BRCA1/2 pathogenic variant carriers and those with HRD (homologous recombination deficiency). Concerns over toxicity and costs have led to the search for predictive biomarkers. We present an updated systematic review, expanding on a previous ESMO review on PARP inhibitor biomarkers. METHODS Following ESMO's 2020 review protocol, we extended our search to March 31, 2023, including PubMed and clinical trial data. We also reviewed the reference lists of review articles. We conducted a meta-analysis using a random-effects model to evaluate hazard ratios and assess the predictive potential of biomarkers and the effectiveness of PARP inhibitors in survival. RESULTS We found 375 articles, 103 of which were included after screening (62 primary research, 41 reviews). HRD remained the primary biomarker (95%), particularly BRCA1/2 variants (77%). In the non-HRD category, six articles (10%) introduced innovative biomarkers, including ADP-ribosylation, HOXA9 promoter methylation, patient-derived organoids, KELIM, and SLFN11. DISCUSSION Prospective assessment of real-time homologous recombination repair via nuclear RAD51 levels shows promise but needs validation. Emerging biomarkers like ADP-ribosylation, HOXA9 promoter methylation, patient-derived organoids, KELIM, and SLFN11 offer potential but require large-scale validation.
Collapse
Affiliation(s)
- Ying-Wen Wang
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynaecology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Isaac Allen
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Yvonne Walburga Joko-Fru
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Gong T, Jiang J, Uthayopas K, Bornman MSR, Gheybi K, Stricker PD, Weischenfeldt J, Mutambirwa SBA, Jaratlerdsiri W, Hayes VM. Rare pathogenic structural variants show potential to enhance prostate cancer germline testing for African men. Nat Commun 2025; 16:2400. [PMID: 40064858 PMCID: PMC11893795 DOI: 10.1038/s41467-025-57312-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Prostate cancer (PCa) is highly heritable, with men of African ancestry at greatest risk and associated lethality. Lack of representation in genomic data means germline testing guidelines exclude for Africans. Established that structural variations (SVs) are major contributors to human disease and prostate tumourigenesis, their role is under-appreciated in familial and therapeutic testing. Utilising clinico-methodologically matched deep-sequenced whole-genome data for 113 African versus 57 European PCa patients, we interrogate 42,966 high-quality germline SVs using a best-fit pathogenicity prediction workflow. We identify 15 potentially pathogenic SVs representing 12.4% African and 7.0% European patients, of which 72% and 86% met germline testing standard-of-care recommendations, respectively. Notable African-specific loss-of-function gene candidates include DNA damage repair MLH1 and BARD1 and tumour suppressors FOXP1, WASF1 and RB1. Representing only a fraction of the vast African diaspora, this study raises considerations with respect to the contribution of kilo-to-mega-base rare variants to PCa pathogenicity and African-associated disparity.
Collapse
Affiliation(s)
- Tingting Gong
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Jue Jiang
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Korawich Uthayopas
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia
| | - M S Riana Bornman
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Kazzem Gheybi
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia
| | | | - Joachim Weischenfeldt
- Finsen Laboratory, Rigshospitalet, DK-2200, Copenhagen, Denmark
- Biotech Research & Innovation Centre, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Shingai B A Mutambirwa
- Department of Urology, Sefako Makgatho Health Science University, Dr George Mukhari Academic Hospital, Medunsa, Ga-Rankuwa, South Africa
| | - Weerachai Jaratlerdsiri
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Vanessa M Hayes
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia.
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.
- Manchester Cancer Research Centre, University of Manchester, Manchester, M20 4GJ, UK.
| |
Collapse
|
22
|
Pinto Á, Domínguez M, Gómez-Iturriaga A, Rodriguez-Vida A, Vallejo-Casas JA, Castro E. The role of radium-223 in the evolving treatment landscape of metastatic castration-resistant prostate cancer: A narrative review. Crit Rev Oncol Hematol 2025; 210:104678. [PMID: 40058740 DOI: 10.1016/j.critrevonc.2025.104678] [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: 12/18/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The treatment of metastatic castration-resistant prostate cancer (mCRPC) has been rapidly evolving over the last two decades. The advent of new androgen receptor pathway inhibitors (ARPIs) such as abiraterone acetate or enzalutamide marks a great advance for treating mCRPC patientd in the pre- and post-docetaxel settings. The subsequent approval of ARPIs in early stages-i.e., metastatic hormone-sensitive (mHSPC) or nonmetastatic CRPC-led to a realignment of subsequent treatment choices upon progression to mCRPC, given the possibility of cross-resistance between ARPIs. Therapies with mechanisms of action different from those of ARPIs are now the focus of new treatment developments. Also, this anomalous situation brings the focus back to well-known treatments currently used later in the treatment sequence. This is the case of radium-223 which, when administered with enzalutamide, has recently been shown to prolong radiographic progression-free survival vs. enzalutamide alone in the first line in asymptomatic or mildly symptomatic patients with no known visceral metastases. In this narrative review, we summarize the treatment landscape for mCRPC, both from a historical and practical point of view, to understand the new potential of radium-223 as a treatment option in this setting.
Collapse
Affiliation(s)
- Álvaro Pinto
- Medical Oncology Department, Hospital Universitario La Paz, Madrid, Spain.
| | - Mario Domínguez
- Urology Department. Hospital Universitario Marqués de Valdecilla, Instituto de Investigación de Valdecilla (IDIVAL), Santander, Spain
| | - Alfonso Gómez-Iturriaga
- Radiation Oncology Department, Cruces University Hospital, Biobizkaia Health Research Institute, Basque Country University (UPV/EHU), Bilbao, Spain
| | | | | | - Elena Castro
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
23
|
Lawaczeck L, Rüdiger A, Hennenlotter J, Hammes J, Spingler V, Walz S, Erne E, Tsaur I, Rausch S. Impact of interdisciplinary tumor boards (ITB) and personalized treatment on survival outcomes in metastatic castration-resistant prostate cancer. J Cancer Res Clin Oncol 2025; 151:101. [PMID: 40047924 PMCID: PMC11885382 DOI: 10.1007/s00432-025-06135-8] [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: 01/01/2025] [Accepted: 02/06/2025] [Indexed: 03/09/2025]
Abstract
PURPOSE Interdisciplinary tumor boards (ITB) are essential in optimizing treatment recommendations for metastatic castration-resistant prostate cancer (mCRPC) by incorporating oncology guidelines, clinical trials, and patient-specific factors to ensure individualized care. This study examines clinical parameters that influence ITB recommendations, evaluates their adherence to guidelines, and assesses their impact on patient survival. METHODS In a retrospective analysis, data from 187 mCRPC patients discussed at an ITB in a tertiary care center in 2018 were evaluated. Patient- and disease-specific factors were correlated with adherence to National Comprehensive Cancer Network® (NCCN®) guidelines and overall survival (OS). The impact of clinical parameters on survival outcomes was assessed through univariate and multivariate analyses. RESULTS The median patient age was 72.8 years, with a median prostate-specific antigen (PSA) level of 65.0 ng/ml. Guideline-compliant recommendations were given in 42.9% of cases, while 57.1% received individualized recommendations. Clinical trial eligibility was noted in 24.8% of patients. Individualized ITB recommendations were associated with significantly longer OS (38.3 vs. 21.2 months, p = 0.03). Shorter OS correlated with renal impairment (p = 0.007), symptomatic metastases (p < 0.0001), and visceral metastases (p < 0.0001). Limitations include the retrospective design, lack of follow-up on therapy adherence, and absence of progression-free survival (PFS) data. CONCLUSION ITB discussions improve survival in mCRPC patients, mainly due to personalized approaches and better access to clinical trials. Visceral and symptomatic metastases as well as renal impairment are risk factors for reduced OS, emphasizing the need for careful management of these high-risk patients. The results support the expanded use of ITB to improve mCRPC treatment outcomes.
Collapse
Affiliation(s)
- Laura Lawaczeck
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Anna Rüdiger
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Joël Hammes
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Valentina Spingler
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Simon Walz
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Eva Erne
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Igor Tsaur
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Steffen Rausch
- Department of Urology, Klinik Für Urologie, Eberhard-Karls-University, Universitätsklinik Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany.
| |
Collapse
|
24
|
Fadlullah MZH, Nix D, Herberts C, Maurice-Dror C, Wyatt AW, Schmidt B, Fairbourn B, Tan AC, Wang L, Kohli M. Multi-gene risk score for prediction of clinical outcomes in treatment-naïve metastatic castrate-resistant prostate cancer. JNCI Cancer Spectr 2025; 9:pkaf025. [PMID: 40036789 PMCID: PMC11954629 DOI: 10.1093/jncics/pkaf025] [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: 11/14/2024] [Revised: 01/16/2025] [Accepted: 02/15/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND To determine the performance of a multi-gene copy number variation (MG-CNV) risk score in metastatic tissue and plasma biospecimens from treatment-naïve metastatic castration-resistant prostate cancer (mCRPC) patients for prediction of clinical outcomes. METHODS The mCRPC tissue and plasma cell-free DNA (cfDNA) biospecimen sequencing results obtained from publicly accessed cohorts in dbGaP, cBioPortal, and an institutional mCRPC cohort were used to develop a MG-CNV risk score derived from gains in AR, MYC, COL22A1, PIK3CA, PIK3CB, NOTCH1 and losses in TMPRSS2, NCOR1, ZBTB16, TP53, NKX3-1 in independent cohorts for determining overall survival (OS), progression-free survival (PFS) to first-line androgen receptor pathway inhibitors (ARPIs). The range of the risk scores for each cohort was dichotomized into "high-risk" and "low-risk" groups and association with OS/PFS determined. Univariate and multivariable Cox proportional hazards regressions were applied for survival analyses (P < .05 for statistical significance). RESULTS Of 1137 metastatic tissue-plasma biospecimens across all cohorts, 699/1137 were treatment-naive mCRPC (235/699 metastatic tissue; 464/699 plasma-cfDNA), and 311/1137 were matched tissue-cfDNA pairs. In multivariable analysis, the MG-CNV risk score derived from metastatic tissue or in cfDNA was statistically significantly associated with OS with high score associated with short survival (hazard ratio = 2.65, confidence interval = 1.99 to 3.51; P = 1.35-11) and shorter PFS to ARPIs (median PFS of 7.8 months) compared with 14 months in patients with low-risk score. CONCLUSIONS A molecular risk score in treatment-naïve mCRPC state obtained either in metastatic tissue or cfDNA predicts clinical survival outcomes and offers a tumor biology-based tool to design biomarker-based enrichment clinical trials.
Collapse
Affiliation(s)
- Muhammad Zaki Hidayatullah Fadlullah
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Informatics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - David Nix
- Department of Biomedical Informatics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Bogdana Schmidt
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Brayden Fairbourn
- Department of Internal Medicine, Spencer Fox Eccles School of Medicine, Salt Lake City, UT, United States
| | - Aik-Choon Tan
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Informatics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Liang Wang
- Department of Tumor Biology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Manish Kohli
- Division of Oncology, Department of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
25
|
Muzzana M, Broggini M, Damia G. The Landscape of PARP Inhibitors in Solid Cancers. Onco Targets Ther 2025; 18:297-317. [PMID: 40051775 PMCID: PMC11884256 DOI: 10.2147/ott.s499226] [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: 10/15/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
PARP inhibitors are a class of agents that have shown significant preclinical activity in models defective in homologous recombination (HR). The identification of synthetic lethality between HR defects and PARP inhibition led to several clinical trials in tumors with known HR defects (initially mutations in BRCA1/2 genes and subsequently in other genes involved in HR). These studies demonstrated significant responses in breast and ovarian cancers, which are known to have a significant proportion of patients with HR defects. Since the approval of the first PARP inhibitor (PARPi), olaparib, several other inhibitors have been developed, expanding the armamentarium available to clinicians in this setting. The positive results obtained in breast and ovarian cancer have expanded the use of PARPi in other solid tumors with HR defects, including prostate and pancreatic cancer in which these defects have been identified. The clinical trials have demonstrated responses to PARPi which are now also available for the subset of patients with prostate and pancreatic cancer with HR defects. This review summarizes the results obtained in solid tumors with PARPi and their potential use when combined with other agents, including immune checkpoint inhibitors that are likely to further increase the survival of these patients which still needs a dramatic improvement.
Collapse
Affiliation(s)
- Marta Muzzana
- Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Massimo Broggini
- Experimental Oncology Department, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giovanna Damia
- Experimental Oncology Department, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
26
|
Suvac A, Ashton J, Bristow RG. Tumour hypoxia in driving genomic instability and tumour evolution. Nat Rev Cancer 2025; 25:167-188. [PMID: 39875616 DOI: 10.1038/s41568-024-00781-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/30/2025]
Abstract
Intratumour hypoxia is a feature of all heterogenous solid tumours. Increased levels or subregions of tumour hypoxia are associated with an adverse clinical prognosis, particularly when this co-occurs with genomic instability. Experimental evidence points to the acquisition of DNA and chromosomal alterations in proliferating hypoxic cells secondary to inhibition of DNA repair pathways such as homologous recombination, base excision repair and mismatch repair. Cell adaptation and selection in repair-deficient cells give rise to a model whereby novel single-nucleotide mutations, structural variants and copy number alterations coexist with altered mitotic control to drive chromosomal instability and aneuploidy. Whole-genome sequencing studies support the concept that hypoxia is a critical microenvironmental cofactor alongside the driver mutations in MYC, BCL2, TP53 and PTEN in determining clonal and subclonal evolution in multiple tumour types. We propose that the hypoxic tumour microenvironment selects for unstable tumour clones which survive, propagate and metastasize under reduced immune surveillance. These aggressive features of hypoxic tumour cells underpin resistance to local and systemic therapies and unfavourable outcomes for patients with cancer. Possible ways to counter the effects of hypoxia to block tumour evolution and improve treatment outcomes are described.
Collapse
Affiliation(s)
- Alexandru Suvac
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jack Ashton
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert G Bristow
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
27
|
Bastian MB, Sieben M, Blickle A, Burgard C, Speicher T, Bartholomä M, Schaefer-Schuler A, Maus S, Ezziddin S, Rosar F. Safety of PSMA radioligand therapy in mCRPC patients with preexisting moderate to severe thrombocytopenia. Eur J Nucl Med Mol Imaging 2025; 52:1271-1277. [PMID: 39625666 PMCID: PMC11839894 DOI: 10.1007/s00259-024-07006-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/24/2024] [Indexed: 02/20/2025]
Abstract
PURPOSE Aim of this study was to analyze the safety of prostate-specific membrane antigen radioligand therapy (PSMA-RLT) in patients with metastatic castration-resistant prostate cancer (mCRPC) with preexisting moderate to severe thrombocytopenia (CTCAE ≥ 2). MATERIALS AND METHODS Seventeen mCRPC patients with preexisting thrombocytopenia (platelet count < 75 × 109/L) were included in this study. Patients received a median of 3 cycles of [177Lu]Lu-PSMA-617 (range 1-6). The course of platelet cell count was closely monitored within and after the PSMA-RLT and analyzed statistically and according to CTCAE. RESULTS No significant difference in platelet counts was observed between baseline and follow-up after each PSMA-RLT cycle: first cycle (54.18 ± 16.07 at baseline vs. 59.65 ± 39.16 at follow up [in × 109/L], p= 0.834), second cycle (58.56 ± 16.43 vs. 107.1 ± 56.44, p = 0.203), and third cycle (60.38 ± 16.57 vs. 132.1 ± 80.43, p = 0.148), respectively. Similarly, baseline and end of treatment values, irrespective of the number of administered cycles, did not reveal a significant difference (54.18 ± 16.07 vs. 72.06 ± 71.9, p = 0.741). After the end of therapy, irrespective of the number of administered cycles, 29.4% of patients remained stable in terms of CTCAE scoring, 41.2% changed to a higher score and 29.4% improved to a lower score. We observed no critical bleeding events due to thrombocytopenia. CONCLUSION Despite the common consideration of marked preexisting thrombocytopenia as a contraindication for RLT, this study indicates feasibility of PSMA-RLT in patients with preexisting thrombocytopenia of grade ≥ 2, as in our preliminary experience, there was no RLT-induced significant deterioration of platelet cell count. Thus, patients with thrombocytopenia should not be categorically excluded from receiving PSMA-RLT.
Collapse
Affiliation(s)
- Moritz B Bastian
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Maike Sieben
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Arne Blickle
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Caroline Burgard
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Tilman Speicher
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Mark Bartholomä
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Andrea Schaefer-Schuler
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Stephan Maus
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany
| | - Florian Rosar
- Department of Nuclear Medicine, Saarland University, Kirrberger Str., Geb. 50, 66421, Homburg, Germany.
| |
Collapse
|
28
|
Rao M, Merrill M, Troxel M, Chiang S, Momeni-Boroujeni A, Hensley ML, Schram AM. Retrospective Analysis of BRCA-Altered Uterine Sarcoma Treated With Poly(ADP-ribose) Polymerase Inhibitors. JCO Precis Oncol 2025; 9:e2400765. [PMID: 40117531 PMCID: PMC11949232 DOI: 10.1200/po-24-00765] [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: 10/21/2024] [Revised: 12/28/2024] [Accepted: 01/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Uterine sarcomas are rare, aggressive tumors with limited chemotherapy responsiveness. Poly(ADP-ribose) polymerase inhibitors (PARPis) have emerged as targeted therapies for patients with BRCA mutations across multiple cancer types, with anecdotal responses in uterine sarcoma. This retrospective, single-center study aims to describe relevant genomic and clinical features of patients with BRCA-altered uterine sarcoma and the efficacy of PARPis in this population. METHODS Eligible patients included all histopathologically confirmed uterine sarcoma with pathogenic BRCA alterations identified through Memorial Sloan Kettering Cancer Center-integrated mutation profiling of actionable cancer targets, excluding carcinosarcoma. Genomic, pathologic, and treatment information was extracted from the cBioPortal database and chart review. RESULTS Thirty-five patients were identified with uterine sarcoma harboring pathogenic BRCA alterations, including 33 BRCA2 alterations (70% homozygous deletions, 3% structural variants, 27% mutations) and two BRCA1 mutations. Leiomyosarcoma (LMS) was the most common histology (86%). Thirteen patients with uterine LMS were treated with PARPis in the recurrent/metastatic therapy setting (54% combination therapy regimens) with an overall response rate (ORR) of 46% (1 of 6 for PARPi monotherapy, 5 of 7 for PARPi combination regimens), a clinical benefit rate (CBR) of 62%, and a median progression-free survival (PFS) of 13.2 months (range, 1.0-71.9). The median PFS ratio compared with previous systemic therapy was 1.9 (range, 0.4-53.9), and 58% had a PFS ratio of ≥1.3. The median time on PARPi was 14.5 months (range, 1.3-71.9). The ORR for patients with somatic BRCA2 deletions was 60% (n = 6 of 10), with a CBR of 80% (n = 8 of 10). One patient with metastatic disease and progression on previous hormonal and chemotherapy demonstrated a complete response to PARP/PD-L1 inhibitor combination therapy, ongoing for 70+ months. CONCLUSION PARPis demonstrate promising efficacy in patients with uterine LMS with somatic BRCA2 deletions.
Collapse
Affiliation(s)
- Mara Rao
- Rutgers New Jersey Medical School, Newark, NJ
| | | | - Megan Troxel
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sarah Chiang
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Martee L. Hensley
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Alison M. Schram
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
29
|
Obinata D, Yamada Y, Sumiyoshi T, Tanegashima T, Watanabe R, Kobayashi H, Ito D, Urabe F. Recent advances in basic research on prostate cancer: Where we are heading? Int J Urol 2025; 32:219-228. [PMID: 39474871 DOI: 10.1111/iju.15628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/17/2024] [Indexed: 03/21/2025]
Abstract
In the over 80 years since androgens were found to play a pivotal role in prostate cancer (PCa) progression, androgen deprivation therapy (ADT) has been a cornerstone in treating advanced PCa. Castration-resistant PCa persists, however, with some of these tumors evolving to androgen receptor (AR)-independent forms like neuroendocrine PCa. The development of novel diagnostic and therapeutic approaches to PCa is therefore crucial. This review provides an overview of recent basic research in PCa, focusing on two main areas: PCa cells and their tumor microenvironments. The first section describes current knowledge on the intricate mechanisms of AR signaling pathways, emphasizing the roles of coactivators and chromatin state alterations in gene regulation. Genomic analyses have revealed recurrent mutations and copy number alterations critical for precision medicine. Liquid biopsy has become a promising tool for real-time tumor monitoring, identifying genetic alterations in circulating-tumor DNA or extracellular vesicles. The second section describes the tumor microenvironment of PCa, highlighting its immunosuppressive landscape and the potential of combining ADT with immunotherapy. Advanced techniques, including single-cell RNA sequencing and spatial transcriptomics offer insights into cellular heterogeneity and interactions within the tumor microenvironment, paving the way for novel therapeutic strategies. Integration of these diverse research areas will provide a comprehensive understanding of the current state and future directions of PCa research, underscoring the importance of personalized medicine and the dynamic nature of cancer treatment strategies.
Collapse
Affiliation(s)
- Daisuke Obinata
- Department of Urology, Nihon University School of Medicine, Tokyo, Japan
| | - Yasutaka Yamada
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takayuki Sumiyoshi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tokiyoshi Tanegashima
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuta Watanabe
- Department of Urology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroaki Kobayashi
- Department of Urology, National Defense Medical College, Saitama, Japan
| | - Daisuke Ito
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Shi B, Du M, Chen Z. Advances in tumor immunotherapy targeting macrophages. Expert Rev Clin Immunol 2025; 21:259-276. [PMID: 39636579 DOI: 10.1080/1744666x.2024.2438721] [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: 04/08/2024] [Revised: 11/03/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION In recent years, immunotherapy has shown significant therapeutic potential in patients with advanced tumors. However, only a small number of individuals benefit, mainly due to the tumor microenvironment (TME), which provides conditions for the development of tumors. Macrophages in TME, known as tumor-associated macrophages (TAM), are mainly divided into M1 anti-tumor and M2 pro-tumor phenotypes, which play a regulatory role in various stages of tumorigenesis, promote tumorigenesis and metastasis, and cause immunotherapy resistance. AREAS COVERED This review focuses on research strategies and preclinical/clinical research progress in translating TAM into antitumor phenotype by referring to the PubMed database for five years. These include small molecule chemotherapy drug development, metabolic regulation, gene editing, physical stimulation, nanotechnology-mediated combination therapy strategies, and chimeric antigen receptor-based immunotherapy. EXPERT OPINION It is necessary to explore the surface-specific receptors and cell signaling pathways of TAM further to improve the specificity and targeting of drugs and to strengthen research in the field of probes that can monitor changes in TAM in real time. In addition, the physical stimulation polarization strategy has the advantages of being noninvasive, economical, and stable and will have excellent clinical transformation value in the future.
Collapse
Affiliation(s)
- Binrui Shi
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Medical imaging, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
31
|
Hintelmann K, Böckelmann L. [Olaparib for high-risk biochemically recurrent prostate cancer following prostatectomy]. Strahlenther Onkol 2025; 201:343-345. [PMID: 39690262 DOI: 10.1007/s00066-024-02350-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Affiliation(s)
- Katharina Hintelmann
- Klinik für Strahlentherapie und Ambulanzzentrum der UKE GmbH, Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland.
| | - Lukas Böckelmann
- Klinik für Strahlentherapie und Ambulanzzentrum der UKE GmbH, Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland
| |
Collapse
|
32
|
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
|
33
|
Beauchamp L, Indulkar S, Erak E, Salimian M, Matoso A. Tissue-Based Biomarkers Important for Prognostication and Management of Genitourinary Tumors, Including Surrogate Markers of Genomic Alterations. Surg Pathol Clin 2025; 18:175-189. [PMID: 39890303 DOI: 10.1016/j.path.2024.10.002] [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
A better understanding of the molecular alterations that underlie urologic malignancies and advances in targeted therapies has impacted classification, prognostication, and treatment. In bladder tumors, these advances include the development of antibody-drug conjugates targeting nectin-4 and Trop-2, as well as human epidermal growth factor receptor 2 and immunotherapy. In prostate cancer, assessment of the percentage of Gleason pattern 4, presence of cribriform glands, and molecular alterations, including PTEN and mismatch repair protein loss, have become standard for clinical care. In renal malignancies, alterations in TSC1/2, mammalian target of rapamycin, anaplastic lymphoma kinase, and other genes impact classification and therapeutic decisions.
Collapse
Affiliation(s)
- Leonie Beauchamp
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Shreeya Indulkar
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Eric Erak
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Andres Matoso
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA; Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA.
| |
Collapse
|
34
|
Steenbock O, Paffenholz P, Rieger C, Heidenreich J, Pfister D, von Brandenstein M, Heidenreich A. [Next generation sequencing (NGS)-based molecular panel analysis for metastatic prostate cancer: how often can we detect druggable mutations? : NGS for metastatic adenocarcinoma of the prostate]. UROLOGIE (HEIDELBERG, GERMANY) 2025; 64:256-268. [PMID: 39836208 PMCID: PMC11893637 DOI: 10.1007/s00120-024-02493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Prostate cancer guidelines recommend molecular analysis of biomaterial following resistance to first-line systemic therapy in order to identify druggable mutations. We report on our results of molecular analysis of tissue specimens via next generation sequencing (NGS) in men with metastatic castration resistant prostate cancer (mCRPC). PATIENTS AND METHODS In all, 311 mCRPC patients underwent NGS analysis from biopsy samples of progressive metastatic lesions or archival radical prostatectomy specimens. NGS analysis was either performed using a panel of 18 prostate cancer-specific amplicons or via the TS0500 panel. RESULTS Of the 311 biopsies, 299 (96%) revealed sufficient DNA content for NGS analysis independent on the specimen origin. Biopsies were taken from prostate (31%), lymph nodes (26%), visceral (17%) or osseous (18%) metastases. In 223 (75%) and 76 (25%) patients activating/inhibiting and no mutations were identified, respectively. Most frequently, mutations of HRD genes including a positive HRD score and p53 were identified in 22% of patients each. About 50% of HRD gene mutations were pathogenic and treatment with PARP inhibitors was initiated. Although the majority of p53 alterations were inactivating mutations, 3 patients demonstrated gain-of-function mutations resulting in an inactivation of ATM. Activating androgen receptor mutations and inactivating PTEN mutations were identified in 42 (14%) and 24 (8%) patients, respectively. Specific AR mutations resulted in a switch of hormonal therapy. Mutations of mismatch repair deficiency genes/MSI high were identified in 5 patients resulting in the administration of pembrolizumab. Addition of the TSO 500 panel identified additional mutations in 4.5% of patients and only 2% of the total cohort would have benefitted from this large panel with the identification of druggable mutations. CONCLUSION Druggable mutations were identified in one third of mCRPC patients using an 18 amplicon-panel analyzed via NGS. Based on our data, molecular analysis of AR mutations or mutations of the HRD genes should be performed following progression after first-line hormonal therapy. A more extensive molecular analysis seems to be useful following progression to the standard sequential hormonal, cytotoxicand radioligand therapies. Use of the expensive TSO 500 panel seems to be of additional therapeutic value in only a minority of patients.
Collapse
Affiliation(s)
- Olivia Steenbock
- Klinik für Urologie, Uro-Onkologie, roboter-assistierte und spezielle urologische Chirurgie, Uniklinik Köln, Kerpener Str. 62, 50927, Köln, Deutschland
| | - Pia Paffenholz
- Klinik für Urologie, Uro-Onkologie, roboter-assistierte und spezielle urologische Chirurgie, Uniklinik Köln, Kerpener Str. 62, 50927, Köln, Deutschland
| | - Constantin Rieger
- Klinik für Urologie, Uro-Onkologie, roboter-assistierte und spezielle urologische Chirurgie, Uniklinik Köln, Kerpener Str. 62, 50927, Köln, Deutschland
| | - Julian Heidenreich
- Klinik für Urologie, Uro-Onkologie, roboter-assistierte und spezielle urologische Chirurgie, Uniklinik Köln, Kerpener Str. 62, 50927, Köln, Deutschland
| | - David Pfister
- Klinik für Urologie, Uro-Onkologie, roboter-assistierte und spezielle urologische Chirurgie, Uniklinik Köln, Kerpener Str. 62, 50927, Köln, Deutschland
| | - Melanie von Brandenstein
- Klinik für Urologie, Uro-Onkologie, roboter-assistierte und spezielle urologische Chirurgie, Uniklinik Köln, Kerpener Str. 62, 50927, Köln, Deutschland
| | - Axel Heidenreich
- Klinik für Urologie, Uro-Onkologie, roboter-assistierte und spezielle urologische Chirurgie, Uniklinik Köln, Kerpener Str. 62, 50927, Köln, Deutschland.
- Institut für Pathologie, Uniklinik Köln, Köln, Deutschland.
- Klink für Urologie, Medizinische Universität Wien, Wien, Österreich.
| |
Collapse
|
35
|
Hage Chehade C, Gebrael G, Sayegh N, Ozay ZI, Narang A, Crispino T, Golan T, Litton JK, Swami U, Moore KN, Agarwal N. A pan-tumor review of the role of poly(adenosine diphosphate ribose) polymerase inhibitors. CA Cancer J Clin 2025; 75:141-167. [PMID: 39791278 PMCID: PMC11929130 DOI: 10.3322/caac.21870] [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: 09/09/2024] [Accepted: 10/03/2024] [Indexed: 01/12/2025] Open
Abstract
Poly(adenosine diphosphate ribose) polymerase (PARP) inhibitors, such as olaparib, talazoparib, rucaparib, and niraparib, comprise a therapeutic class that targets PARP proteins involved in DNA repair. Cancer cells with homologous recombination repair defects, particularly BRCA alterations, display enhanced sensitivity to these agents because of synthetic lethality induced by PARP inhibitors. These agents have significantly improved survival outcomes across various malignancies, initially gaining regulatory approval in ovarian cancer and subsequently in breast, pancreatic, and prostate cancers in different indications. This review offers a comprehensive clinical overview of PARP inhibitor approvals, emphasizing their efficacy across different cancers based on landmark phase 3 clinical trials.
Collapse
Affiliation(s)
- Chadi Hage Chehade
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Nicolas Sayegh
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zeynep Irem Ozay
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Arshit Narang
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Tony Crispino
- UsTOO Prostate Cancer Support and Education Las Vegas Chapter, Las Vegas, Nevada, USA
| | - Talia Golan
- Division of Medical Oncology, Sheba Medical Center, Tel Aviv Medical University, Tel Aviv, Israel
| | - Jennifer K Litton
- Division of Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Kathleen N Moore
- Division of Gynecologic Oncology, Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
36
|
Singh S, Kumar R, Tripathi S, Salahuddin, Mazumder A, Singh N. Fused and Substituted Piperazines as Anticancer Agents: A Review. Chem Biol Drug Des 2025; 105:e70077. [PMID: 40047279 DOI: 10.1111/cbdd.70077] [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: 01/05/2025] [Revised: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
Cancer is an abnormal and uncontrolled proliferation of normal cells. The availability of safer anticancer drugs with exceptional selectivity for healthy cells and great efficacy against various cancer forms continues to be a significant obstacle. The piperazine moiety is used as the building block of several molecules and is reported to have the ability to inhibit the cell cycle (G1/S phase), inhibit angiogenesis, and interact with DNA. Piperazine also has a flexible binding feature that allows it to interact with a variety of biological targets, which makes it effective against cancers. As there is a continuous need to obtain an anticancer drug with improved efficacy and fewer side effects, the piperazine derivatives attract the attention of researchers. This review highlights the recently reported methods of synthesis of fused/substituted piperazines, structure-activity relationship, and interactions with targets/receptors as anticancer agents. Thus, the presented review will help medicinal chemists in designing anticancer molecules with piperazines.
Collapse
Affiliation(s)
- Saumya Singh
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Rajnish Kumar
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Shrishti Tripathi
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Salahuddin
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Avijit Mazumder
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Nardev Singh
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
37
|
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:jnumed.124.267730. [PMID: 40015917 DOI: 10.2967/jnumed.124.267730] [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: 12/04/2024] [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
|
38
|
Poon DMC, Cheung WSK, Chiu PKF, Chung DHS, Kung JBT, Lam DCM, Leung AKC, Ng ACF, O’Sullivan JM, Teoh JYC, Wu PY, Wu SKK, Kwong PWK. Treatment of metastatic castration-resistant prostate cancer: review of current evidence and synthesis of expert opinions on radioligand therapy. Front Oncol 2025; 15:1530580. [PMID: 40071082 PMCID: PMC11893367 DOI: 10.3389/fonc.2025.1530580] [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: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Background Despite the boom in the development of cancer management in the last decade, most patients with metastatic prostate cancer (PCa) eventually progress to metastatic castration-resistant PCa (mCRPC) and often require multiple lines of treatment. The treatment landscape of mCRPC has evolved rapidly in recent years, introducing various types of systemic therapies, including taxane-based chemotherapy, androgen receptor pathway inhibitors, bone-targeted radionuclides (e.g., radium-223), immune checkpoint inhibitors, poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitors, and radioligand therapies (RLTs) [e.g., a prostate-specific membrane antigen (PSMA) ligand labelled with 177Lu]. Methods To help clinicians navigate the increasingly complex treatment landscape of mCRPC, this article reviews the evidence on different therapeutic regimens from pivotal trials. In addition, it reports on the results of a questionnaire developed and distributed by the Hong Kong Society of Uro-Oncology (HKSUO), with the aim of collecting the perspectives of specialists experienced in the treatment of advanced PCa in Hong Kong with regard to the clinical application of RLT, primarily [177Lu]Lu-PSMA-617/analogue therapy. Results A total of 43 questionnaire respondents (including clinical oncologists, urologists, nuclear medicine specialists, and medical oncologists) voted on 27 consensus questions divided into eight sections. Consensus or strong consensus (correspondingly ≥75% or ≥90% acceptance for an answer option) was reached for 10 questions. Subsequently, a panel of 13 local and overseas experts coordinated by the HKSUO discussed the voting results and provided further insights into certain questions. Conclusion The literature review, the voting results of the questionnaire, and the expert opinions are expected to facilitate better understanding of recent therapeutic advancements and the role of novel RLTs in the treatment of mCRPC among clinicians.
Collapse
Affiliation(s)
- Darren M. C. Poon
- Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK
Pao Centre for Cancer, Hong Kong Cancer Institute, The Chinese University of Hong
Kong, Hong Kong, Hong Kong SAR, China
- Comprehensive Oncology Centre, Hong Kong Sanatorium and Hospital,
Hong Kong, Hong Kong SAR, China
| | - William S. K. Cheung
- Department of Nuclear Medicine & PET, Hong Kong Sanatorium and
Hospital, Hong Kong, Hong Kong SAR, China
| | - Peter K. F. Chiu
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong
Kong, Hong Kong, Hong Kong SAR, China
| | - Daniel H. S. Chung
- Department of Clinical Oncology, Queen Elizabeth Hospital,
Hong Kong, Hong Kong SAR, China
| | - John B. T. Kung
- Nuclear Medicine Unit, Department of Diagnostic and Interventional Radiology, Queen
Elizabeth Hospital, Hong Kong, Hong Kong SAR, China
| | - Daisy C. M. Lam
- Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales
Hospital, Hong Kong, Hong Kong SAR, China
| | | | - Anthony C. F. Ng
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong
Kong, Hong Kong, Hong Kong SAR, China
| | - Joe M. O’Sullivan
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Hong Kong, Hong Kong SAR, China
| | - Jeremy Y. C. Teoh
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong
Kong, Hong Kong, Hong Kong SAR, China
| | - Philip Y. Wu
- Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong, Hong Kong SAR, China
| | - Sam K. K. Wu
- Department of Nuclear Medicine & PET, Hong Kong Sanatorium and
Hospital, Hong Kong, Hong Kong SAR, China
| | | |
Collapse
|
39
|
Sweatman E, Bayley R, Selemane R, Higgs MR. SETD1A-dependent EME1 transcription drives PARPi sensitivity in HR deficient tumour cells. Br J Cancer 2025:10.1038/s41416-025-02963-0. [PMID: 39994444 DOI: 10.1038/s41416-025-02963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Cells deficient in DNA repair factors breast cancer susceptibility 1/2 (BRCA1/2) or ataxia-telangiectasia mutated (ATM) are sensitive to poly-ADP ribose polymerase (PARP) inhibitors. Building on our previous findings, we asked how the lysine methyltransferase SETD1A contributed to PARP inhibitor-mediated cell death in these contexts and determined the mechanisms responsible. METHODS We used cervical, breast, lung and ovarian cancer cells bearing mutations in BRCA1 or ATM and depleted SETD1A using siRNA or CRISPR/Cas9. We assessed the effects of the PARPi Olaparib on cell viability, homologous recombination, and DNA repair. We assessed underlying transcriptional perturbations using RNAseq. We used The Cancer Genomics Atlas (TCGA) and DepMap to investigate patient survival and cancer cell characteristics. RESULTS Loss of SETD1A from both BRCA1-deficient and ATM-deficient cancer cells was associated with resistance to Olaparib, explained by partial restoration of homologous recombination. Mechanistically, SETD1A-dependent transcription of the crossover junction endonuclease EME1 correlated with sensitivity to Olaparib in these cells. Accordingly, when SETD1A or EME1 was lost, BRCA1 or ATM-mutated cells became resistant to Olaparib, and homologous recombination was partially restored. CONCLUSIONS Loss of SETD1A or EME1 drives cellular resistance to Olaparib in certain genetic contexts and may help explain why patients develop resistance to PARP inhibitors in the clinic.
Collapse
Affiliation(s)
- Ellie Sweatman
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Rachel Bayley
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Richad Selemane
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Martin R Higgs
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
40
|
Taniguchi H, Ikeda J, Masuo Y, Kinoshita H. The impact of the number of prior androgen receptor pathway inhibitors before cabazitaxel treatment in patients with metastatic castration-resistant prostate cancer. Jpn J Clin Oncol 2025:hyaf034. [PMID: 39987487 DOI: 10.1093/jjco/hyaf034] [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: 12/12/2024] [Accepted: 02/09/2025] [Indexed: 02/25/2025] Open
Abstract
OBJECTIVE Cabazitaxel (CAZ) has been shown to prolong overall survival (OS) in patients with metastatic castration-resistant prostate cancer (mCRPC) following docetaxel treatment. However, the impact of the number of prior androgen receptor pathway inhibitors (ARPIs) on CAZ efficacy remains unclear. This study aimed to analyze the effectiveness of CAZ based on the number of prior ARPIs administered before CAZ treatment. METHODS A retrospective review was conducted on mCRPC patients who received CAZ. The differences in CAZ efficacy based on the number of prior ARPIs were evaluated and prognostic factors for prostate-specific antigen (PSA) progression-free survival (PFS) and OS were analyzed. RESULTS A total of 59 patients were categorized into three groups: 12 patients with no prior ARPI, 26 with one prior ARPI, and 21 with two or more prior ARPIs. The median number of CAZ cycles and relative dose intensity were 5% and 60%, respectively. No significant differences were observed in PSA response or PFS among the three groups. Although OS from the first CAZ administration did not significantly differ among the groups, the OS from mCRPC diagnosis was shorter in the group with no prior ARPI. Multivariate analysis identified a time to mCRPC diagnosis of less than 11 months and low serum hemoglobin levels before CAZ administration as significant prognostic factors of poor OS following CAZ treatment. CONCLUSIONS PSA response, PFS, and OS after CAZ administration did not significantly differ based on the number of prior ARPIs. CAZ efficacy appears consistent regardless of the number of prior ARPI sequences.
Collapse
Affiliation(s)
- Hisanori Taniguchi
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| | - Junichi Ikeda
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| | - Yuki Masuo
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| |
Collapse
|
41
|
Witz A, Dardare J, Betz M, Michel C, Husson M, Gilson P, Merlin JL, Harlé A. Homologous recombination deficiency (HRD) testing landscape: clinical applications and technical validation for routine diagnostics. Biomark Res 2025; 13:31. [PMID: 39985088 PMCID: PMC11846297 DOI: 10.1186/s40364-025-00740-y] [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: 06/24/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
The use of poly(ADP-ribose) polymerase inhibitors (PARPi) revolutionized the treatment of BRCA-mutated cancers. Identifying patients exhibiting homologous recombination deficiency (HRD) has been proved useful to predict PARPi efficacy. However, obtaining HRD status remains an arduous task due to its evolution over the time. This causes HRD status to become obsolete when obtained from genomic scars, rendering PARPi ineffective for these patients. Only two HRD tests are currently FDA-approved, both based on genomic scars detection and BRCA mutations testing. Nevertheless, new technologies for obtaining an increasingly reliable HRD status continue to evolve. Application of these tests in clinical practice is an additional challenge due to the need for lower costs and shorter time to results delay.In this review, we describe the currently available methods for HRD testing, including the methodologies and corresponding tests for assessing HRD status, and discuss the clinical routine application of these tests and their technical validation.
Collapse
Affiliation(s)
- Andréa Witz
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France.
| | - Julie Dardare
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Margaux Betz
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Cassandra Michel
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Marie Husson
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Pauline Gilson
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Jean-Louis Merlin
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Alexandre Harlé
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
42
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XHF, Zheng J. Invasion and metastasis in cancer: molecular insights and therapeutic targets. Signal Transduct Target Ther 2025; 10:57. [PMID: 39979279 PMCID: PMC11842613 DOI: 10.1038/s41392-025-02148-4] [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: 05/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
The progression of malignant tumors leads to the development of secondary tumors in various organs, including bones, the brain, liver, and lungs. This metastatic process severely impacts the prognosis of patients, significantly affecting their quality of life and survival rates. Research efforts have consistently focused on the intricate mechanisms underlying this process and the corresponding clinical management strategies. Consequently, a comprehensive understanding of the biological foundations of tumor metastasis, identification of pivotal signaling pathways, and systematic evaluation of existing and emerging therapeutic strategies are paramount to enhancing the overall diagnostic and treatment capabilities for metastatic tumors. However, current research is primarily focused on metastasis within specific cancer types, leaving significant gaps in our understanding of the complex metastatic cascade, organ-specific tropism mechanisms, and the development of targeted treatments. In this study, we examine the sequential processes of tumor metastasis, elucidate the underlying mechanisms driving organ-tropic metastasis, and systematically analyze therapeutic strategies for metastatic tumors, including those tailored to specific organ involvement. Subsequently, we synthesize the most recent advances in emerging therapeutic technologies for tumor metastasis and analyze the challenges and opportunities encountered in clinical research pertaining to bone metastasis. Our objective is to offer insights that can inform future research and clinical practice in this crucial field.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Graduate School of Biomedical Science, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lijun Deng
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
43
|
Yu EY, Rumble RB, Agarwal N, Cheng HH, Eggener SE, Bitting RL, Beltran H, Giri VN, Spratt D, Mahal B, Lu K, Crispino T, Trabulsi EJ. Germline and Somatic Genomic Testing for Metastatic Prostate Cancer: ASCO Guideline. J Clin Oncol 2025; 43:748-758. [PMID: 39787437 DOI: 10.1200/jco-24-02608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE To evaluate evidence on germline and somatic genomic testing for patients with metastatic prostate cancer and provide recommendations. METHODS A systematic review by a multidisciplinary panel with patient representation was conducted. The PubMed database was searched from January 2018 to May 2024. Articles were selected for inclusion if they reported on patients with metastatic prostate cancer who received a germline or somatic genomic test and/or made comparisons between those tests, reported detection rates, prognostic information, or treatment implications. RESULTS A total of 1,713 papers were identified in the literature search. After applying the eligibility criteria, 14 remained: eight systematic reviews and six clinical trials. RECOMMENDATIONS Patients with metastatic prostate cancer should undergo both germline and somatic DNA sequencing using panel-based assays. These tests can guide the use of poly(ADP-ribose) polymerase inhibitors, which have a survival benefit in metastatic castration-resistant prostate cancer. In addition, germline testing may have screening implications for additional cancers for patients and cascade testing implications for family members. The data supporting when to perform repeat testing and optimal tissue type to use (eg, primary tumor v metastatic biopsy versus circulating tumor DNA [ctDNA] testing) are more limited, but this panel recommends considering retesting in patients whose results were previously negative or uninformative, and to consider either a metastatic biopsy or ctDNA when a significant change in clinical status occurs. Next-generation genomic sequencing findings that are associated with prognostic only (and not predictive) value should not be used to guide treatment outside of a clinical trial.Additional information is available at www.asco.org/genitourinary-cancer-guidelines.
Collapse
Affiliation(s)
- Evan Y Yu
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Neeraj Agarwal
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT
| | - Heather H Cheng
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | | | - Veda N Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, CT
| | | | | | - Kevin Lu
- Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Tony Crispino
- Patient Representative, Chapter President UsTOO Las Vegas, Prostate Cancer Education and Support, Las Vegas, NV
| | - Edouard J Trabulsi
- Jefferson Einstein Medical Center, Sidney Kimmel Cancer Center of Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
44
|
Mitchell J, Camacho N, Shea P, Stopsack KH, Joseph V, Burren OS, Dhindsa RS, Nag A, Berchuck JE, O'Neill A, Abbasi A, Zoghbi AW, Alegre-Díaz J, Kuri-Morales P, Berumen J, Tapia-Conyer R, Emberson J, Torres JM, Collins R, Wang Q, Goldstein D, Matakidou A, Haefliger C, Anderson-Dring L, March R, Jobanputra V, Dougherty B, Carss K, Petrovski S, Kantoff PW, Offit K, Mucci LA, Pomerantz M, Fabre MA. Assessing the contribution of rare protein-coding germline variants to prostate cancer risk and severity in 37,184 cases. Nat Commun 2025; 16:1779. [PMID: 39971927 PMCID: PMC11839991 DOI: 10.1038/s41467-025-56944-1] [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: 05/10/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
To assess the contribution of rare coding germline genetic variants to prostate cancer risk and severity, we perform here a meta-analysis of 37,184 prostate cancer cases and 331,329 male controls from five cohorts with germline whole exome or genome sequencing data, and one cohort with imputed array data. At the gene level, our case-control collapsing analysis confirms associations between rare damaging variants in four genes and increased prostate cancer risk: SAMHD1, BRCA2 and ATM at the study-wide significance level (P < 1×10-8), and CHEK2 at the suggestive threshold (P < 2.6×10-6). Our case-only analysis, reveals that rare damaging variants in AOX1 are associated with more aggressive disease (OR = 2.60 [1.75-3.83], P = 1.35×10-6), as well as confirming the role of BRCA2 in determining disease severity. At the single-variant level, our study reveals that a rare missense variant in TERT is associated with substantially reduced prostate cancer risk (OR = 0.13 [0.07-0.25], P = 4.67×10-10), and confirms rare non-synonymous variants in a further three genes associated with reduced risk (ANO7, SPDL1, AR) and in three with increased risk (HOXB13, CHEK2, BIK). Altogether, this work provides deeper insights into the genetic architecture and biological basis of prostate cancer risk and severity, with potential implications for clinical risk prediction and therapeutic strategies.
Collapse
Affiliation(s)
- Jonathan Mitchell
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| | - Niedzica Camacho
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Patrick Shea
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Konrad H Stopsack
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Vijai Joseph
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Oliver S Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Dhindsa
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Abhishek Nag
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Amanda O'Neill
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ali Abbasi
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Anthony W Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jesus Alegre-Díaz
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
| | - Pablo Kuri-Morales
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Tecnológico, Monterrey, Nuevo León, Mexico
| | - Jaime Berumen
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
| | - Roberto Tapia-Conyer
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
| | - Jonathan Emberson
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jason M Torres
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - David Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Athena Matakidou
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Carolina Haefliger
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Lauren Anderson-Dring
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ruth March
- Precision Medicine and Biosamples, R&D Oncology, AstraZeneca, Dublin, Ireland
| | - Vaidehi Jobanputra
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | | | - Keren Carss
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Convergent Therapeutics, Cambridge, MA, USA
| | - Kenneth Offit
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- American Cancer Society, Atlanta, GA, USA
| | | | - Margarete A Fabre
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
45
|
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
|
46
|
Francolini G, Bertini N, Di Cataldo V, Garlatti P, Aquilano M, Caini S, Bruni A, Ingrosso G, D'angelillo RM, Tagliaferri L, Augugliaro M, Triggiani L, Parisi S, Timon G, Arcidiacono F, Marvaso G, Jereczek-Fossa BA, Lancia A, Franzese C, Alongi F, Simontacchi G, Greto D, Bonomo P, Loi M, Frosini G, Burchini L, Desideri I, Meattini I, Valicenti RK, Livi L. Impact of stereotactic body radiotherapy after progression in castrate resistant prostate cancer patients undergoing first line abiraterone treatment. A subgroup analysis from ARTO trial (NCT03449719). Prostate Cancer Prostatic Dis 2025:10.1038/s41391-025-00950-3. [PMID: 39972049 DOI: 10.1038/s41391-025-00950-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND ARTO trial was a phase II randomized trial suggesting the benefit of a concomitant treatment strategy including Abiraterone acetate plus predisone (AAP) and stereotactic body radiotherapy (SBRT) in oligometastatic castrate resistant prostate cancer (omCRPC). The object of the current analysis is to explore whether the benefit provided by SBRT to AAP is maintained at later stages of disease after oligoprogression METHODS: Patients enrolled in ARTO trial in whom a first progression event was reported were divided in two groups according to the treatment approach received, regardless of the initial randomization. After first progression event, Patients in Group A received SBRT on oligoprogressive disease, while patients in group B received second line systemic treatment. Palliative RT was not considered for the purpose of this analysis. Progression-Free survival (PFS) 1 and 2 were defined as time between AAP start and first progression event and time between first and second progression event, death or last follow up, (whichever came first), respectively. Cox regression analysis was performed to compare PFS1 + PFS2 in patients in group A vs Group B. Kaplan-Meier analysis was performed to compare overall survival between the two groups RESULTS: Median PFS1 + PFS2 was 45.9 months vs. not reached in group A (n = 43) vs Group B (n = 20), respectively (HR 0.63, 95% CI 0.17-2.33, p value 0.489), no significant difference was detected. Median OS was not reached in neither of the two arms of treatment, with a non-significant trend in favour of the experimental arm (HR 0.50, 95% CI 0.14-1.78, p = 0.284) CONCLUSIONS: Results from the present analysis show that SBRT after progression may be a viable and feasible option for omCRPC after progression if compared to second line systemic therapy.
Collapse
Affiliation(s)
- Giulio Francolini
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy.
| | - Niccolò Bertini
- Department of Biomedical, Experimental and Clinical sciences "Mario Serio", University of Florence, Florence, Italy
| | - Vanessa Di Cataldo
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Pietro Garlatti
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Michele Aquilano
- Cyberknife Unit, Istituto Fiorentino di Cura ed Assistenza, IFCA, Florence, Italy
| | - Saverio Caini
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Alessio Bruni
- Radiation Oncology Unit, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Gianluca Ingrosso
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rolando Maria D'angelillo
- Radiation Oncology, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Luca Tagliaferri
- UOC di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario "A.Gemelli" IRCCS, Rome, Italy
| | - Matteo Augugliaro
- Unit of Radiotherapy, Azienda USL-IRCCS di Reggio Emilia, Reggio emilia, Italy
| | - Luca Triggiani
- Università degli Studi di Brescia, Department of Radiation Oncology, Brescia University, Brescia, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Sciences and Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Giorgia Timon
- Unit of Radiotherapy, Azienda USL-IRCCS di Reggio Emilia, Reggio emilia, Italy
| | | | - Giulia Marvaso
- Division of Radiation Oncology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Andrea Lancia
- Department of Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ciro Franzese
- Department of Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Filippo Alongi
- Department of Advanced Radiation Oncology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Gabriele Simontacchi
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Daniela Greto
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Pierluigi Bonomo
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Mauro Loi
- Radiation Oncology Unit, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Giulio Frosini
- Department of Biomedical, Experimental and Clinical sciences "Mario Serio", University of Florence, Florence, Italy
| | - Luca Burchini
- Department of Biomedical, Experimental and Clinical sciences "Mario Serio", University of Florence, Florence, Italy
| | - Isacco Desideri
- Department of Biomedical, Experimental and Clinical sciences "Mario Serio", University of Florence, Florence, Italy
| | - Icro Meattini
- Department of Biomedical, Experimental and Clinical sciences "Mario Serio", University of Florence, Florence, Italy
| | | | - Lorenzo Livi
- Department of Biomedical, Experimental and Clinical sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
47
|
Borbiev T, Babcock K, Sinopole K, Chesnut GT, Petrovics G. Ancestry-Specific DNA Damage Repair Gene Mutations and Prostate Cancer. Cancers (Basel) 2025; 17:682. [PMID: 40002276 PMCID: PMC11853348 DOI: 10.3390/cancers17040682] [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: 01/14/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
This review is intended to reflect the currently available literature on both clinically significant germline mutations in DNA damage repair (DDR) genes as well as the importance of ancestral diversity in the pathogenesis of prostate cancer (PCa). The second most prevalent cancer worldwide in men is PCa, causing significant morbidity and mortality in its advanced stage. Emerging data highlight the substantial role of germline mutations of DDR genes in PCa pathogenesis, especially in progression to aggressive forms of the disease. Germline genetic testing is recognized as a necessary tool for efficient, individualized patient care. NCCR guidelines recommend inquiring about the family history of PCa and known germline variants and, if indicated, proceeding with germline multigene testing followed by post-test genetic counseling. Depending on the germline mutations in HR repair genes or in MMR genes, specific treatment options may provide clinical benefit. We will discuss specific germline mutations that are involved in PCa progression and prognosis in racially diverse populations.
Collapse
Affiliation(s)
- Talaibek Borbiev
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.B.); (G.T.C.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Kevin Babcock
- Internal Medicine, Alexander T. Augusta Military Medicine Center, Fort Belvoir, VA 22060, USA;
| | - Kayleigh Sinopole
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA;
| | - Gregory T. Chesnut
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.B.); (G.T.C.)
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.B.); (G.T.C.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| |
Collapse
|
48
|
Arce-Gallego S, Cresta Morgado P, Delgado-Serrano L, Simonetti S, Gonzalez M, Romero-Lozano P, Marmolejo D, Morales-Barrera R, Arnau GM, Semidey ME, Aguilar D, Cordoba-Terreros S, Mast R, de Albert M, Planas J, Cuadras M, Maldonado X, Suarez C, Casanova-Salas I, Figols M, Cros S, Mas A, Nonell L, Dienstmann R, Nuciforo P, Vivancos A, Llop-Guevara A, Carles J, Serra V, Mateo J. Homologous recombination repair status in metastatic prostate cancer by next-generation sequencing and functional immunofluorescence. Cell Rep Med 2025; 6:101937. [PMID: 39914385 DOI: 10.1016/j.xcrm.2025.101937] [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: 07/21/2024] [Revised: 09/19/2024] [Accepted: 01/10/2025] [Indexed: 02/21/2025]
Abstract
Metastatic prostate cancer (mPC) is enriched for homologous recombination repair (HRR) gene alterations, which have prognostic and predictive value. Routine clinical implementation of next-generation sequencing (NGS) is still limited. We investigated the association between genomic and functional loss of HRR, using NGS and RAD51 immunofluorescence (RAD51-IF) in 219 primary or metastatic biopsies from 187 patients with stage IV prostate cancer. NGS showed frequent genomic alterations in TP53 (40%), AR (15%), PTEN (14%), FOXA1 (12%), MYC (10%), BRCA2 (9%), ATM (8%), and BRCA1 (2%). We pursued RAD51-IF in 206 samples; of those, 139/206 (67%) were evaluable. 21% of samples had RAD51-low score compatible with HRR deficiency (HRD). RAD51-IF showed high sensitivity (71%) and specificity (86%) for BRCA1/2 alterations. Patients with RAD51-low scores experienced longer progression-free survival (PFS) on poly(ADP-ribose) polymerase inhibitors (PARPi) or platinum chemotherapy. RAD51-IF is feasible in routine clinical samples from patients with mPC and is associated with clinically relevant HRR gene alterations.
Collapse
Affiliation(s)
- Sara Arce-Gallego
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pablo Cresta Morgado
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Oncology Data Science (OdysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Luisa Delgado-Serrano
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Sara Simonetti
- Molecular Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Macarena Gonzalez
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paula Romero-Lozano
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - David Marmolejo
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rafael Morales-Barrera
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Gisela Mir Arnau
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Daniel Aguilar
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Richard Mast
- Department of Radiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Matias de Albert
- Department of Radiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Jacques Planas
- Department of Urology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Mercè Cuadras
- Department of Urology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Xavier Maldonado
- Department of Radiation Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Cristina Suarez
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Irene Casanova-Salas
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Mariona Figols
- Department of Medical Oncology, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Spain
| | - Sara Cros
- Department of Medical Oncology, Granollers Hospital, Granollers, Spain
| | - Alba Mas
- Bioinformatics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Lara Nonell
- Bioinformatics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (OdysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; University of Vic - Central University of Catalonia, Vic, Spain
| | - Paolo Nuciforo
- Molecular Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Alba Llop-Guevara
- Experimental Therapeutics, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Joan Carles
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Violeta Serra
- Experimental Therapeutics, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Joaquin Mateo
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
49
|
Ofner H, Kramer G, Shariat SF, Hassler MR. TP53 Deficiency in the Natural History of Prostate Cancer. Cancers (Basel) 2025; 17:645. [PMID: 40002239 PMCID: PMC11853097 DOI: 10.3390/cancers17040645] [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: 12/24/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Prostate cancer remains a leading cause of cancer-related mortality in men, with advanced stages posing significant treatment challenges due to high morbidity and mortality. Among genetic alterations, TP53 mutations are among the most prevalent in cancers and are strongly associated with poor clinical outcomes and therapeutic resistance. This review investigates the role of TP53 mutations in prostate cancer progression, prognosis, and therapeutic development. A comprehensive analysis of preclinical and clinical studies was conducted to elucidate the molecular mechanisms, clinical implications, and potential therapeutic approaches associated with TP53 alterations in prostate cancer. TP53 mutations are highly prevalent in advanced stages, contributing to genomic instability, aggressive tumor phenotypes, and resistance to standard treatments. Emerging evidence supports the utility of liquid biopsy techniques, such as circulating tumor DNA analysis, for detecting TP53 mutations, providing prognostic value and facilitating early intervention strategies. Novel therapeutic approaches targeting TP53 have shown promise in preclinical settings, but their clinical efficacy requires further validation. Overall, TP53 mutations represent a critical biomarker for disease progression and therapeutic response in prostate cancer. Advances in detection methods and targeted therapies hold significant potential to improve outcomes for patients with TP53-mutated prostate cancer. Further research is essential to integrate TP53-based strategies into routine clinical practice.
Collapse
Affiliation(s)
- Heidemarie Ofner
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
| | - Shahrokh F. Shariat
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Urology, Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic
- Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
- Karl Landsteiner Institute of Urology and Andrology, 1090 Vienna, Austria
| | - Melanie R. Hassler
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
| |
Collapse
|
50
|
Khalid F, Bodla ZH, Gaddameedi SR, Macasaet R, Yagnik K, Niaz Z, Fish PN, Du D, Shah S. Evidence-Based Recommendations on the Use of Immunotherapies and Monoclonal Antibodies in the Treatment of Male Reproductive Cancers. Curr Oncol 2025; 32:108. [PMID: 39996908 PMCID: PMC11854063 DOI: 10.3390/curroncol32020108] [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: 03/24/2024] [Revised: 11/14/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
The incidence of male reproductive cancers, including prostate, testicular, and penile cancers, has risen in recent years, raising important health concerns. Prostate cancer is the second leading cause of cancer-related mortality in men, while penile cancer, though rare, typically affects men over 60. Testicular cancer, with a lifetime risk of about 0.4% in men, is most common among adolescents and young adults, decreasing sharply after the age of 40. Traditional treatments include chemotherapy, radiation, surgery, and combinations thereof, but advancements in immunotherapy and monoclonal antibodies are showing promising results, particularly for genitourinary cancers. These therapies, targeting immune checkpoints and tumor-specific antigens, are gaining traction as effective alternatives for resistant cases. This review provides evidence-based recommendations on current and emerging immunotherapy and monoclonal antibody treatments for male reproductive cancers, highlighting future directions to optimize patient outcomes.
Collapse
Affiliation(s)
- Farhan Khalid
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA (S.R.G.); (K.Y.)
| | - Zubair Hassan Bodla
- Department of Internal medicine, Graduate Medical Education, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Sai Rakshith Gaddameedi
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA (S.R.G.); (K.Y.)
| | - Raymart Macasaet
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA (S.R.G.); (K.Y.)
| | - Karan Yagnik
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA (S.R.G.); (K.Y.)
| | - Zahra Niaz
- Department of Medicine, Al-Aleem Medical College, Lahore 54000, Pakistan
| | - Peter N. Fish
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA (S.R.G.); (K.Y.)
| | - Doantrang Du
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA (S.R.G.); (K.Y.)
| | - Shazia Shah
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA (S.R.G.); (K.Y.)
| |
Collapse
|