1
|
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] [MESH Headings] [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
|
2
|
Munzur AD, Bacon JVW, Fishbein F, Herberts C, Donnellan G, Bernales CQ, Parekh K, Vandekerkhove G, Müller DC, Liao YJR, Stephenson M, Nappi L, Khalaf D, Maurice-Dror C, Chi KN, Eigl BJ, Kollmannsberger C, Soleimani M, Wyatt AW. Clonal hematopoiesis in metastatic urothelial and renal cell carcinoma. NPJ Precis Oncol 2025; 9:177. [PMID: 40514379 PMCID: PMC12166084 DOI: 10.1038/s41698-025-00965-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Clonal hematopoiesis (CH) is an age-related expansion of white blood cell (WBC) progenitors linked to risk of hematological malignancy. Patients with cancer have increased CH prevalence compared to healthy populations, but the characteristics and relevance of CH in advanced urological cancers are unknown. We interrogated CH and circulating tumor DNA (ctDNA) in 299 patients with metastatic urothelial or renal cell carcinoma using error-corrected targeted sequencing of matched WBC DNA and plasma cell-free DNA (cfDNA). 73% of patients carried CH variants at ≥0.25% allele frequency, with 13% exhibiting large CH populations marked by variants ≥10%. CH presence, clone size, and genotype did not impact patient survival. However, CH variants frequently affected solid cancer driver genes and were not individually discriminable from ctDNA variants based on cfDNA features including fragment length. In contrast, matched WBC DNA sequencing to ≥25% of cfDNA depth sufficiently resolved CH from ctDNA variants. Serial profiling revealed ctDNA and CH temporal dynamics including treatment-related expansion of PPM1D-mutated CH clones following platinum chemotherapy. Our data reveal the molecular landscape of CH in urological cancers and suggest that CH interferes in clinical ctDNA genotyping. We urge test providers to comprehensively filter CH from ctDNA results using matched WBC sequencing and propose a cost-effective framework for its integration into existing plasma-only assays.
Collapse
Affiliation(s)
- Aslı D Munzur
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jack V W Bacon
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Francine Fishbein
- Department of Internal Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Gráinne Donnellan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Cecily Q Bernales
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Karan Parekh
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Gillian Vandekerkhove
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - David C Müller
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Urology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Yi Jou Ruby Liao
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Maria Stephenson
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lucia Nappi
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Daniel Khalaf
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | | | - Kim N Chi
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Bernhard J Eigl
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Christian Kollmannsberger
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Maryam Soleimani
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
- Clinical Cancer Genomics Program and Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Wu Y, Wang Y, Wang Y, Qiu H, Yuan X, Xiong H, Zou Y. Exploring synthetic lethality in cancer therapy: CRISPR-Cas9 technology offers new hope. Biochim Biophys Acta Rev Cancer 2025; 1880:189370. [PMID: 40516634 DOI: 10.1016/j.bbcan.2025.189370] [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: 07/04/2024] [Revised: 06/06/2025] [Accepted: 06/08/2025] [Indexed: 06/16/2025]
Abstract
Synthetic lethality (SL) is a breakthrough concept in cancer therapy that describes a scenario in which the simultaneous inactivation of two genes leads to cell death, whereas inactivation of either gene alone does not. The rise of clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated nuclease 9 (Cas9) technology has provided a new tool for exploring this phenomenon, enabling genome editing and screening. This review evaluates the advancements achieved by CRISPR technology in identifying novel therapeutic targets and comprehending the processes of drug resistance using the concept of SL in cancer cells. This review explores the fundamental concept of SL and its application in cancer therapy, highlighting how the CRISPR-Cas9 system functions and how CRISPR-based screening can be leveraged to identify synthetic lethal genes and investigate the mechanisms of drug resistance. We summarize important research in related fields from recent years, demonstrating the role of CRISPR screening in revealing cancer cellular pathways and identifying new drug targets. We also summarize the clinical trials of related drugs currently underway, and anticipate that with the continuous development of CRISPR technology, its integration with cancer genetics and immuno-oncology will bring new hope to patients with drug-resistant cancers.
Collapse
Affiliation(s)
- Yuqi Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yali Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanbin Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yanmei Zou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Hushmandi K, Farahani N, Einollahi B, Salimimoghadam S, Alimohammadi M, Liang L, Liu L, Sethi G. Deciphering molecular pathways in urological cancers: a gateway to precision therapeutics. J Adv Res 2025:S2090-1232(25)00395-9. [PMID: 40516913 DOI: 10.1016/j.jare.2025.06.009] [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: 03/03/2025] [Revised: 06/02/2025] [Accepted: 06/05/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Urological cancers, including prostate, kidney, bladder, testicular, and penile cancers, pose a significant health challenge, particularly in their metastatic stages. Surgical interventions remain fundamental, but recent advancements in medical therapies like chemotherapy, immunotherapy, and targeted therapies have shown promise in improving patient outcomes. AIM OF REVIEW This review aims to explore the current landscape of targeted therapies in urological cancers, focusing on the role of key signaling pathways such as phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt), mechanistic (mammalian) target of rapamycin (mTOR), growth factor-related mechanisms, c-Mesenchymal-epithelial transition factor (c-Met)/ hepatocyte growth factor (HGF), programmed cell death protein 1 and its ligand programmed death-ligand 1 (PD-1/PD-L1), and steroid hormone receptor pathways in tumor progression and therapeutic resistance. Key scientific concepts of review Dysregulation of pathways like PI3K/Akt and mTOR contributes to tumorigenesis, metastasis, and resistance to treatment, underscoring their relevance as therapeutic targets. Tyrosine kinase inhibitors and immune checkpoint inhibitors have demonstrated efficacy but face challenges such as intrinsic resistance and treatment-related toxicities. Integrating insights from signaling pathway research with clinical practice holds potential for developing more effective treatment paradigms, enhancing the efficacy of targeted therapies, and improving survival rates for patients with urological cancers.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran,Iran
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore 117600 Singapore, Singapore.
| |
Collapse
|
5
|
Wosny M, Aeppli S, Fischer S, Peres T, Rothermundt C, Hastings J. A Bayesian Network Meta-analysis of Systemic Treatments for Metastatic Castration-Resistant Prostate Cancer in First- and Subsequent Lines. Target Oncol 2025:10.1007/s11523-025-01148-2. [PMID: 40493311 DOI: 10.1007/s11523-025-01148-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2025] [Indexed: 06/12/2025]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) presents a challenge for clinicians in determining the optimal treatment sequence because of the lack of direct head-to-head comparisons, which is further complicated by the now-widespread use of androgen receptor pathway inhibitors (ARPIs) in metastatic hormone-sensitive prostate cancer (mHSPC). OBJECTIVE This study is a Bayesian network meta-analysis (NMA) intended to provide a comprehensive evaluation and comparison of the efficacy of mCRPC treatments across different treatment lines. PATIENTS AND METHODS We performed a systematic search of ClinicalTrials.gov, extracted information, assessed the risk of bias, and reconstructed missing outcomes. We performed an NMA to evaluate treatment efficacy for overall survival (OS) and progression-free survival (PFS) in first and subsequent lines. The study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) NMA guidelines and was registered with PROSPERO (CRD42024499607). RESULTS The NMA included 43 trials with 33,494 patients. ARPI-based therapies, particularly in combination with poly(ADP-ribose) polymerase inhibitors, demonstrated the most significant benefits for OS and PFS in first-line mCRPC treatment, followed by chemotherapy regimens. However, ARPI re-treatment showed limited effectiveness in subsequent lines, leading to weaker OS and PFS benefits. CONCLUSIONS This NMA highlights the superiority of ARPI-based therapies and chemotherapies as first-line options for mCRPC while emphasizing the need for treatment class switching after ARPI failure. To refine treatment sequencing and enable precision care, future research should integrate individual participant data to better address patient-level heterogeneity and identify biomarkers for personalized therapy.
Collapse
Affiliation(s)
- Marie Wosny
- School of Medicine, University of St. Gallen (HSG), St. Jakob-Strasse 21, 9000, St. Gallen, Switzerland.
- Institute for Implementation Science in Health Care, University of Zurich (UZH), Zurich, Switzerland.
| | - Stefanie Aeppli
- Department of Medical Oncology and Hematology, HOCH Health Ostschweiz, Kantonsspital St. Gallen (KSSG), St. Gallen, Switzerland
| | - Stefanie Fischer
- Department of Medical Oncology and Hematology, HOCH Health Ostschweiz, Kantonsspital St. Gallen (KSSG), St. Gallen, Switzerland
| | - Tobias Peres
- Department of Medical Oncology and Hematology, HOCH Health Ostschweiz, Kantonsspital St. Gallen (KSSG), St. Gallen, Switzerland
| | - Christian Rothermundt
- Department of Medical Oncology and Cancer Center, Luzerner Kantonsspital (LUKS), Lucerne, Switzerland
| | - Janna Hastings
- School of Medicine, University of St. Gallen (HSG), St. Jakob-Strasse 21, 9000, St. Gallen, Switzerland
- Institute for Implementation Science in Health Care, University of Zurich (UZH), Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| |
Collapse
|
6
|
Zacchi F, Abida W, Antonarakis ES, Bryce AH, Castro E, Cheng HH, Shandhu S, Mateo J. Recent and Future Developments in the Use of Poly (ADP-ribose) Polymerase Inhibitors for Prostate Cancer. Eur Urol Oncol 2025; 8:818-828. [PMID: 39638687 DOI: 10.1016/j.euo.2024.11.011] [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: 10/01/2024] [Revised: 10/30/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND OBJECTIVE Advanced prostate cancer (PCa) is enriched for alterations in DNA damage repair genes; poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) are a class of drugs that have demonstrated effectiveness in PCa, particularly in tumors with alterations in BRCA1/2 and other homologous recombination repair (HRR) genes, acting through a synthetic lethal mechanism. To prevent or delay drug resistance, and to expand the patient population that can benefit from this class of drug, combination treatment strategies have been developed in preclinical and clinical studies. METHODS This review examines the latest developments in clinical trials testing PARPi for advanced PCa and their emerging role in earlier disease settings. Furthermore, it discusses the critical role of careful patient selection and identification of additional biomarkers to enhance treatment efficacy. KEY FINDINGS AND LIMITATIONS Two PARPi (olaparib and rucaparib) have been approved as monotherapy in metastatic castration-resistant PCa, thereby establishing the first biomarker-guided drug indications in PCa. Several combinations of PARPi with androgen receptor pathway inhibitors have now also been approved. Anemia and fatigue are the main adverse events associated with this drug class in clinical trials; gastrointestinal toxicities are common but usually manageble. CONCLUSIONS AND CLINICAL IMPLICATIONS PARPi are active against PCa with HRR mutations, especially in those with germline or somatic BRCA1/2 mutations. There is still a need to further optimize patient stratification strategies, particularly for combination approaches. Future research should focus on refining predictive biomarkers, improving treatment delivery strategies, and exploring the potential benefits of PARPi in earlier stages of the disease. PATIENT SUMMARY Here, we summarize the results from clinical trials testing different poly (ADP-ribose) polymerase inhibitors (PARPi), a novel targeted drug class, in prostate cancer. Overall, the data from these trials confirm the efficacy of this drug class in those metastatic prostate cancers that show specific gene alterations, such as mutations in the BRCA1/2 genes. Several studies combining PARPi with other standard drugs for prostate cancer suggest that there may be efficacy in larger patient populations, but some of these data still need validation in longer follow-up analyses.
Collapse
Affiliation(s)
- Francesca Zacchi
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emmanuel S Antonarakis
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN, USA
| | - Alan H Bryce
- Department of Medical Oncology and Developmental Therapeutics, City of Hope, Goodyear, AZ, USA
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Heather H Cheng
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shahneen Shandhu
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Joaquin Mateo
- Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
| |
Collapse
|
7
|
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] [MESH Headings] [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
|
8
|
Shore N, Nielsen SM, Esplin ED, Antonarakis ES, Barata PC, Beer TM, Beltran H, Bryce A, Cookson MS, Crawford ED, Dorff TB, George DJ, Heath EI, Helfand BT, Hussain M, Mckay RR, Morgans AK, Morris MJ, Paller CJ, Ross AE, Sartor O, Shen J, Sieber P, Smith MR, Wise DR, Armstrong AJ. Implementation of Universal Germline Genetic Testing Into Standard of Care for Patients With Prostate Cancer: The Time Is Now. JCO Oncol Pract 2025; 21:747-753. [PMID: 39700441 PMCID: PMC12162241 DOI: 10.1200/op-24-00626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/25/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
Indications for and implications of germline genetic testing (GGT) in patients with prostate cancer have expanded over the past decade, particularly related to precision therapies and management. GGT has become the standard of care for many cancers such as breast, ovarian, colorectal, pancreatic, and metastatic prostate cancer, and it is imperative that patients be offered timely and equitable access to testing as it can inform patient-physician shared decision making for management of the current cancer as well as anticipatory guidance for disease progression. Additionally, GGT guides screening for and prevention of secondary malignancies for the patient and cascade testing for at-risk family members. Here, we present data supporting the notion that clinicians should offer all patients with prostate cancer the opportunity to undergo comprehensive GGT for pathogenic germline variants known to be associated with familial cancer and/or known to have implications for treatment and management.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | | | | | | | | | - Tomasz M. Beer
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Exact Sciences Corporation, Madison, WI
| | | | - Alan Bryce
- City of Hope Cancer Center, Goodyear, AZ
| | - Michael S. Cookson
- Stephenson Cancer Center, OU Health, The University of Oklahoma, Oklahoma City, OK
| | | | | | | | | | - Brian T. Helfand
- NorthShore University HealthSystem/Endeavor Health, Evanston, IL
| | - Maha Hussain
- Northwestern Feinberg School of Medicine, Chicago, IL
| | | | | | | | - Channing J. Paller
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | - John Shen
- UCLA David Geffen School of Medicine, Los Angeles, CA
| | | | | | - David R. Wise
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | | |
Collapse
|
9
|
Overbeek JK, Guchelaar NA, Mohmaed Ali MI, Sark M, Hovenier C, Kievit W, Ligtenberg MJ, Ottevanger PB, Bloemendal HJ, Koolen SL, Mathijssen RH, Boere IA, Huitema AD, Sonke GS, Opdam FL, ter Heine R, van Erp NP. Pharmacokinetic boosting of olaparib: Study protocol of a multicentre, open-label, randomised, non-inferiority trial (PROACTIVE-B). Contemp Clin Trials Commun 2025; 45:101477. [PMID: 40248173 PMCID: PMC12005849 DOI: 10.1016/j.conctc.2025.101477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/09/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
Background Pharmacokinetic (PK) boosting is the intentional use of a drug-drug interaction to enhance systemic drug exposure. PK boosting of the anticancer drug olaparib, a CYP3A-substrate, has the potential to reduce PK variability, side effects and financial burden associated with this drug. After establishing adequate pharmacokinetic exposure with boosting in the PROACTIVE-A study, the PROACTIVE-B study is designed to evaluate non-inferiority for both efficacy and toxicity of the boosted therapy compared to the standard monotherapy of olaparib. Methods The PROACTIVE-B study is a nationwide, multicentre, prospective, randomized, non-inferiority trial. A total of 142 patients (128 patients with BRCA+, high-grade, FIGO III/IV ovarian cancer who receive olaparib as maintenance therapy; 14 patients with other approved indications for olaparib) who start olaparib treatment in line with the drug label will be randomized between the standard monotherapy of olaparib 300 mg twice daily (BID) and the boosted therapy of olaparib 100 mg BID with cobicistat 150 mg BID. The co-primary objectives are tolerability (dose reductions due to toxicity), and efficacy (progression-free survival at 12 months) in the ovarian cancer population. Secondary objectives include health status (EQ-5D-5L), patient satisfaction (Cancer Therapy Satisfaction Questionnaire (CTSQ)), and cost effectiveness using the institute for Medical Technology Assessment (iMTA) Productivity Cost Questionnaire (iPCQ) and iMTA Medical Consumption Questionnaire (iMCQ). Discussion PK boosting of olaparib is a potentially valuable strategy to reduce the olaparib dose and the variability in olaparib exposure with fewer side effects. Moreover, the lower costs related to the boosted therapy contribute to a durable and accessible anticancer treatment for all patients. Trial registration The PROACTIVE study has been published at ClinicalTrials.gov under NCT05078671 on October 14, 2021 and at EudraCT under 2021-004032-28 on August 24, 2021.
Collapse
Affiliation(s)
- Joanneke K. Overbeek
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niels A.D. Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ma Ida Mohmaed Ali
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Muriëlle Sark
- Patient Advisory Group, Breast Cancer Research Group and Dutch Breast Cancer Association, Amsterdam, the Netherlands
| | | | - Wietske Kievit
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marjolijn J.L. Ligtenberg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Haiko J. Bloemendal
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Stijn L.W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ron H.J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ingrid A. Boere
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alwin D.R. Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gabe S. Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frans L. Opdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rob ter Heine
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nielka P. van Erp
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
10
|
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
|
11
|
Vázquez-Estévez S, Gallardo E, Fernández-Calvo O, Juan-Fita MJ, Montesa-Pino Á, Lázaro-Quintela M, Anido-Herranz U, González-Del-Alba A. Expert Opinion on Current Treatment Alternatives for Patients With Prostate Cancer Progressing From the Metastatic Hormone-Sensitive Stage to the Castration-Resistant Stage After Receiving Early Treatment Intensification. Clin Genitourin Cancer 2025; 23:102338. [PMID: 40252319 DOI: 10.1016/j.clgc.2025.102338] [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: 12/20/2024] [Revised: 02/25/2025] [Accepted: 03/12/2025] [Indexed: 04/21/2025]
Abstract
For patients with castration-sensitive prostate cancer (mCSPC), treatment intensification with androgen deprivation therapy (ADT) plus new androgen receptor pathway inhibitors (ARPIs) has opened a scenario where no guidance exists to indicate the best treatment after progression to metastatic castration-resistant prostate cancer (mCRPC). Clinical decision-making has become even more complex, with the proven benefit for selected patients of triplet therapy with abiraterone or darolutamide added to the double combination therapy of ADT plus docetaxel. The profile of patients for whom triple therapy would be more beneficial is being defined beyond metastatic disease presentation and volume (eg, poor prognosis features). In October 2023 and October 2024, a panel of eight Spanish medical oncologists with expertise in the management of prostate cancer met to discuss the challenges in treating mCRPC. The scientific evidence was reviewed during this meeting, knowledge and experience were shared, and controversies were discussed until a consensus was reached. This information was collected and turned into a manuscript aimed at helping clinicians determine the optimal treatment sequence after disease progression based on scientific evidence and experts' opinions and consensus. To this end, the profile of mCSPC patients who may have received double or triplet therapy is analyzed, current treatment options are reviewed, and treatment algorithms are proposed. New and expected advancements in this field are also presented.
Collapse
Affiliation(s)
| | - Enrique Gallardo
- Department of Oncology, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Ovidio Fernández-Calvo
- Department of Medical Oncology, Complejo Hospitalario Universitario Ourense, Ourense, Spain
| | - María José Juan-Fita
- Department of Medical Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Álvaro Montesa-Pino
- Department of Medical Oncology, Hospital Regional Universitario de Málaga, Málaga, Spain
| | | | - Urbano Anido-Herranz
- Department of Medical Oncology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | | |
Collapse
|
12
|
Grewal K, Dorff TB, Mukhida SS, Agarwal N, Hahn AW. Advances in Targeted Therapy for Metastatic Prostate Cancer. Curr Treat Options Oncol 2025; 26:465-475. [PMID: 40299225 DOI: 10.1007/s11864-025-01323-7] [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] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
OPINION STATEMENT Over the past few years, treatment for advanced prostate cancer has begun shifting away from a one-size-fits-all approach toward biomarker-based therapies for select groups of patients. This review highlights the role of poly-ADP-ribose-polymerase (PARP) inhibitors in metastatic prostate cancer, emerging strategies to target the androgen receptor (AR), and innovative therapies aimed at cell surface proteins, including radioligand therapies, bispecific T cell engagers, and antibody-drug conjugates. For patients with homologous recombination repair (HRR)-mutated metastatic castration-resistant prostate cancer (CRPC), we favor combining a PARP inhibitor (PARPi) with an AR pathway inhibitor (ARPI), provided they can tolerate a more aggressive treatment strategy. In our opinion, patients with BRCA1 or BRCA2 mutations who are unable to handle combination therapy benefit from PARPi monotherapy. We are enthusiastic about the potential of ongoing clinical trials for new AR-directed therapies, such as AR ligand-directed degraders and CYP11A1 inhibitors, in metastatic CRPC. These treatments are expected to be most beneficial for patients whose cancer continues to rely on AR pathway signaling, suggesting they might also be effective in earlier stages of the disease. Progress in drug development and understanding of protein structures has led to new therapies that target cell surface proteins predominantly found in prostate cancer. We use 177Lu-PSMA-617 for patients with PSMA avid metastatic CRPC who have progressed on an ARPI and a taxane chemotherapy. Additionally, we see promising potential in bispecific T-cell engagers (e.g., STEAP1-CD3 and PSMA-CD3) and novel radioligand therapies, including those utilizing actinium, to target these proteins. These advances show great promise in further enhancing survival for patients with metastatic prostate cancer.
Collapse
Affiliation(s)
- Kabir Grewal
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Tanya B Dorff
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA, USA
| | - Sagar S Mukhida
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeraj Agarwal
- Department of Genitourinary Oncology, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Andrew W Hahn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Goel K, Venkatappa V, Krieger K, Chen D, Sreekumar A, Gassman N. PARP inhibitor response is enhanced in prostate cancer when XRCC1 expression is reduced. NAR Cancer 2025; 7:zcaf015. [PMID: 40271221 PMCID: PMC12015684 DOI: 10.1093/narcan/zcaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
Prostate cancer (PCa) is the second most common cancer worldwide and the fifth leading cause of cancer-related deaths among men. The emergence of metastatic castration-resistant prostate cancer (mCRPC) after androgen deprivation therapy (ADT) exemplifies the complex disease management for PCa. PARP inhibitors (PARPis) are being tested to treat mCRPC in tumors with defective homologous recombination repair (HRR) to address this complexity. However, increasing resistance towards PARPi in HRR-deficient patients and the low percentage of HRR-defective mCRPC patients requires the identification of new genes whose deficiency can be exploited for PARPi treatment. XRCC1 is a DNA repair protein critical in the base excision repair (BER) and single strand break repair (SSBR) pathways. We analyzed PCa patients' cohorts and found that XRCC1 expression varies widely, with many patients showing low XRCC1 expression. We created XRCC1 deficiency in PCa models to examine PARPi sensitivity. XRCC1 loss conferred hypersensitivity to PARPi by promoting the accumulation of DNA double-strand breaks, increasing cell-cycle arrest, and inducing apoptosis. We confirmed that XRCC1 expression correlated with PARPi sensitivity using a doxycycline-inducible system. Therefore, we conclude that XRCC1 expression level predicts response to PARPi, and the clinical utility of PARPi in PCa can extend to low XRCC1 expressing tumors.
Collapse
Affiliation(s)
- Kaveri Goel
- Department of Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Vani Venkatappa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Kimiko L Krieger
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Dongquan Chen
- Division of Preventive Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Arun Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Natalie R Gassman
- Department of Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
14
|
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] [MESH Headings] [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
|
15
|
Loeb S, Vadaparampil ST, Giri VN. Germline testing for prostate cancer: current state and opportunities for enhanced access. EBioMedicine 2025; 116:105705. [PMID: 40398351 PMCID: PMC12148599 DOI: 10.1016/j.ebiom.2025.105705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/16/2025] [Accepted: 04/02/2025] [Indexed: 05/23/2025] Open
Abstract
Germline Testing (GT) for prostate cancer (PCA) is now central to PCA care and hereditary cancer assessment, with a rising role in PCA screening approaches. Guidelines have significantly expanded to include testing patients with metastatic PCA, advanced PCA or with high-risk features, and for males with or without PCA with a strong family cancer history to identify hereditary cancer syndromes for patients and their families. However, the expansion of GT has overwhelmed genetic counselling programs, necessitating the development and evaluation of alternate genetic delivery models. Furthermore, disparities in engagement in PCA GT are of major concern for impacting PCA-related and overall cancer-related outcomes for patients and their families. This review focuses on integrating PCA GT guidelines with implementation strategies and addressing PCA GT disparities to help inform current and future strategies to enhance the benefits of GT across populations.
Collapse
Affiliation(s)
- Stacy Loeb
- Department of Urology and Population Health, New York University Langone Health, New York, NY, USA
| | - Susan T Vadaparampil
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | - Veda N Giri
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
16
|
Naqvi SAA, Riaz IB, Bibi A, Khan MA, Imran M, Khakwani KZR, Raina A, Anjum MU, Cobran EK, Warner JL, Hussain SA, Singh P, Childs DS, Baca SC, Orme JJ, Mateo J, Agarwal N, Gillessen S, Murad MH, Sartor O, Bryce AH. Heterogeneity of the Treatment Effect with PARP Inhibitors in Metastatic Castration-resistant Prostate Cancer: A Living Interactive Systematic Review and Meta-analysis. Eur Urol 2025; 87:626-640. [PMID: 39848867 DOI: 10.1016/j.eururo.2024.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/24/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND AND OBJECTIVE Selection of patients harboring mutations in homologous recombination repair (HRR) genes for treatment with a PARP inhibitor (PARPi) is challenging in metastatic castration-resistant prostate cancer (mCRPC). To gain further insight, we quantitatively assessed the differential efficacy of PARPi therapy among patients with mCRPC and different HRR gene mutations. METHODS This living meta-analysis (LMA) was conducted using the Living Interactive Evidence synthesis framework. We included clinical trials assessing PARPi as monotherapy in pretreated mCRPC or in combination with an androgen receptor pathway inhibitor (ARPI) in treatment-naïve patients. Random-effects meta-analyses were performed for a priori subgroups stratified by HRR status, BRCA status, and each gene. KEY FINDINGS AND LIMITATIONS This first report for our LMA includes 13 trials (4278 patients). Among patients with pretreated mCRPC receiving PARPi monotherapy, the tumor response rate per 100 person-months was numerically higher for patients with BRCA2 (50% prostate-specific antigen response [PSA50%] 3.3; objective response rate [ORR] 3.3), BRCA1 (PSA50% 1.2; ORR 2.0), or PALB2 (PSA50% 3.3; ORR 1.4) alterations than for patients with ATM (PSA50% 0.4; ORR 0.3), CDK12 (PSA50% 0.2; ORR 0.2), or CHEK2 (PSA50% 1.0; ORR 0.7) alterations. Among patients receiving PARPi + ARPI, a significant radiographic progression-free survival benefit was observed in those with BRCA (hazard ratio [HR] 0.28, 95% confidence interval [CI] 0.13-0.62) or CDK12 (HR 0.58, 95% CI 0.35-0.95) alterations, but not in patients with PALB2 (HR 0.53, 95% CI 0.21-1.32), ATM (HR 0.93, 95% CI 0.57-1.53), or CHEK2 (HR 0.92, 95% CI 0.53-1.61) alterations. An overall survival benefit was observed for patients with BRCA alterations (HR 0.47, 95% CI 0.31-0.71) after adjustment for crossover and subsequent therapy, but not for patients with PALB2 (HR 0.33, 95% CI 0.10-1.16), ATM (HR 0.97, 95% CI 0.57-1.67), CDK12 (HR 0.80, 95% CI 0.36-1.78), or CHEK2 (HR 0.81, 95% CI 0.37-1.75) alterations. CONCLUSIONS AND CLINICAL IMPLICATIONS Our LMA delivers information on the effect of PARPi therapy in relation to specific gene alterations in mCRPC via an interactive web platform. The evidence suggests the greatest PARPi benefit in patients with BRCA alterations, a strong signal of benefit in patients with PALB2 or CDK12 alterations, and no benefit in patients with ATM or CHEK2 alterations.
Collapse
Affiliation(s)
| | - Irbaz Bin Riaz
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ, USA.
| | - Arifa Bibi
- Department of Internal Medicine, University of Oklahoma, Oklahoma City, OK, USA
| | - Muhammad Ali Khan
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Manal Imran
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Ammad Raina
- Department of Internal Medicine, Canyon Vista Medical Center, Midwestern University, Sierra Vista, AZ, USA
| | - Muhammad Umair Anjum
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Ewan K Cobran
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ, USA
| | - Jeremy L Warner
- Center for Clinical Cancer Informatics and Data Science, Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Syed A Hussain
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Parminder Singh
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ, USA
| | | | - Sylvan C Baca
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jacob J Orme
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Joaquin Mateo
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Silke Gillessen
- Department of Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland
| | | | - Oliver Sartor
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Alan H Bryce
- Department of Oncology, City of Hope Cancer Center, Goodyear, AZ, USA
| |
Collapse
|
17
|
Geng JH, Li CY, Wu MT, Chen SC, Huang SP. Associations Between Long-Term Exposure to Air Pollutants and Prostate Cancer in a Large Taiwanese Population. Int J Med Sci 2025; 22:2771-2781. [PMID: 40520887 PMCID: PMC12163608 DOI: 10.7150/ijms.109687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/25/2025] [Indexed: 06/18/2025] Open
Abstract
Air pollution is associated with various illnesses including cancers, of which prostate cancer is one of the most prevalent malignancies in men. Emerging evidence has suggested that air pollution is a potential risk factor for prostate cancer. This study aimed to explore the relationship between air pollution and prostate cancer in a Taiwanese population. Using data from the Kaohsiung Medical University Hospital Database, we conducted a case-control study to identify patients with prostate cancer, and matched them by age with individuals without prostate cancer. Environmental pollution indices including particulate matter (PM), nitrogen oxides (NOx), sulfur dioxide (SO2), ozone (O3) and carbon monoxide (CO) were correlated with the patients' addresses using data from the Taiwan Central Air Quality Monitoring Network. The analysis included 3541 prostate cancer patients and 7082 age-matched controls. After adjusting for confounders, conditional logistic regression analysis demonstrated significant associations of prostate cancer with PM2.5 (odds ratio [95% confidence interval]: 1.240 [1.134-1.356]) and CO (odds ratio [95% confidence interval]: 1.105 [1.025-1.192]) at the index date, with similar associations observed for average exposure levels over 1, 2, 3, and 5 years prior to the index date. Furthermore, sensitivity analyses revealed that the odds ratios for combined-risk Z-score exposure at the index date and over these same time periods were 1.029, 1.033, 1.034, 1.034, and 1.033, respectively. These findings suggest that prolonged exposure to multiple air pollutants collectively contributes to prostate cancer risk. Further investigations are needed to validate these findings and explore potential underlying mechanisms.
Collapse
Affiliation(s)
- Jiun-Hung Geng
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Urology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812015, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University 807378, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Ming-Tsang Wu
- Ph.D. Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Szu-Chia Chen
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812015, Taiwan
- Department of Internal Medicine, Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Shu-Pin Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University 807378, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Ph.D. Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Institute of Medical Science and Technology, College of Medicine, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| |
Collapse
|
18
|
Zou Y, Zhang H, Chen P, Tang J, Yang S, Nicot C, Guan Z, Li X, Tang H. Clinical approaches to overcome PARP inhibitor resistance. Mol Cancer 2025; 24:156. [PMID: 40442774 PMCID: PMC12123805 DOI: 10.1186/s12943-025-02355-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
PARP inhibitors have profoundly changed treatment options for cancers with homologous recombination repair defects, especially those carrying BRCA1/2 mutations. However, the development of resistance to these inhibitors presents a significant clinical challenge as it limits long-term effectiveness. This review provides an overview of the current understanding of resistance mechanisms to PARP inhibitors and explores strategies to overcome these challenges. We discuss the basis of synthetic lethality induced by PARP inhibitors and detail diverse resistance mechanisms affecting PARP inhibitors, including homologous recombination restoration, reduced PARP trapping, enhanced drug efflux, and replication fork stabilization. The review then considers clinical approaches to combat resistance, focusing on combination therapies with immune checkpoint inhibitors, DNA damage response inhibitors, and epigenetic drugs. We also highlight ongoing clinical trials and potential biomarkers for predicting treatment response and resistance. The review concludes by outlining future research directions, emphasizing the need for longitudinal studies, advanced resistance monitoring technologies, and the development of novel combination strategies. By tackling PARP inhibitor resistance, this review seeks to aid in the development of more effective cancer therapies, with the potential to improve outcomes for patients with homologous recombination-deficient tumors.
Collapse
Affiliation(s)
- Yutian Zou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Hanqi Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Pangzhou Chen
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Jiayi Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Siwei Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Christophe Nicot
- Department of Pathology and Laboratory Medicine, Rainbow Boulevard, University of Kansas Medical Center, 3901 , Kansas City, KS, 66160, USA
| | - Ziyun Guan
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| | - Xing Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
19
|
Kwan EM, Ng SWS, Tolmeijer SH, Emmett L, Sandhu S, Buteau JP, Iravani A, Joshua AM, Francis RJ, Subhash V, Lee ST, Scott AM, Martin AJ, Stockler MR, Donnellan G, Annala M, Herberts C, Davis ID, Hofman MS, Azad AA, Wyatt AW, TheraP Investigators and the ANZUP Cancer Trials Group. Lutetium-177-PSMA-617 or cabazitaxel in metastatic prostate cancer: circulating tumor DNA analysis of the randomized phase 2 TheraP trial. Nat Med 2025:10.1038/s41591-025-03704-9. [PMID: 40425844 DOI: 10.1038/s41591-025-03704-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/07/2025] [Indexed: 05/29/2025]
Abstract
The prostate-specific membrane antigen (PSMA)-targeted radioligand [¹⁷⁷Lu]Lu-PSMA-617 is a new standard treatment for metastatic castration-resistant prostate cancer (mCRPC), but predictive genomic biomarkers informing its rational use are unknown. We performed detailed dissection of prostate cancer driver genes across 290 serial plasma cell-free DNA samples from 180 molecular imaging-selected patients with mCRPC from the randomized TheraP trial of [¹⁷⁷Lu]Lu-PSMA-617 (n = 97) versus cabazitaxel chemotherapy (n = 83). The primary endpoint was PSA50 biochemical response, with secondary endpoints of progression-free survival (PFS) and overall survival (OS). In this post-hoc biomarker analysis, a low pretreatment circulating tumor DNA (ctDNA) fraction predicted a superior biochemical response (100% versus 58%, P = 0.0067) and PFS (median 14.7 versus 6.0 months; hazard ratio 0.12, P = 2.5 × 10-4) on [¹⁷⁷Lu]Lu-PSMA-617 independent of predictive PSMA-positron emission tomography imaging parameters, although this benefit did not extend to OS. Deleterious PTEN alterations were associated with worse PFS and OS on cabazitaxel, whereas ATM defects were observed in select patients with favorable [¹⁷⁷Lu]Lu-PSMA-617 outcomes. Comparing pretreatment and progression ctDNA revealed population flux but no evidence that alterations in individual mCRPC genes (or FOLH1) are dominant causes of acquired [¹⁷⁷Lu]Lu-PSMA-617 or cabazitaxel resistance. Our results nominate new candidate biomarkers for [¹⁷⁷Lu]Lu-PSMA-617 selection and ultimately expand the mCRPC predictive biomarker repertoire. We anticipate our ctDNA fraction-aware analytical framework will aid future precision management strategies for [¹⁷⁷Lu]Lu-PSMA-617 and other PSMA-targeted therapeutics. ClinicalTrials.gov identifier: NCT03392428 .
Collapse
Affiliation(s)
- Edmond M Kwan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Sarah W S Ng
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sofie H Tolmeijer
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Shahneen Sandhu
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - James P Buteau
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Amir Iravani
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Nuclear Medicine, Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anthony M Joshua
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Medical Oncology, Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Roslyn J Francis
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Vinod Subhash
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Sydney, New South Wales, Australia
| | - Sze-Ting Lee
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Andrew M Scott
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Andrew J Martin
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Martin R Stockler
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Gráinne Donnellan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matti Annala
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian D Davis
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia.
- Cancer Services, Eastern Health, Melbourne, Victoria, Australia.
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Arun A Azad
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada.
| | | |
Collapse
Collaborators
Tim Akhurst, Ramin Alipour, Dale L Bailey, Patricia Banks, Alexis Beaulieu, Louise Campbell, Wei Chua, Megan Crumbaker, Nattakorn Dhiantravan, Kate Ford, Craig Gedye, Jeffrey C Goh, Alex D Guminski, Anis Hamid, Mohammad B Haskali, Rodney J Hicks, Edward Hsiao, Terry Hung, Ian D Kirkwood, Grace Kong, Ailsa Langford, Nicola Lawrence, Jeremy Lewin, Peter Lin, Michael McCarthy, Margaret M McJannett, William McDonald, Kate Moodie, Declan G Murphy, Siobhan Ng, Andrew Nguyen, David A Pattison, David Pook, Izabella Pokorski, Shakher Ramdave, Nisha Rana, Aravind S Ravi Kumar, Andrew D Redfern, Paul Roach, Peter Roselt, Natalie K Rutherford, Javad Saghebi, Geoffrey Schembri, Lavinia Spain, Shalini Subramaniam, Thean Hsiang Tan, Sue Ping Thang, Paul Thomas, Ben Tran, John A Violet, Roslyn Wallace, Andrew Weickhardt, Scott G Williams, Sonia Yip, Alison Y Zhang,
Collapse
|
20
|
Raychaudhuri R, Cheng HH, Gulati R, Schweizer MT, Lin A, Yezefski T, Khan HM, Yu EY, Hawley JE, Nelson PS, Pritchard CC, Montgomery B. Results from a Phase 2 Study of Induction Docetaxel and Carboplatin Followed by Maintenance Rucaparib in the Treatment of Patients with Metastatic Castration-resistant Prostate Cancer with DNA Homologous Recombination Repair Deficiency. Eur Urol Oncol 2025:S2588-9311(25)00123-3. [PMID: 40413129 DOI: 10.1016/j.euo.2025.04.026] [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: 02/14/2025] [Revised: 04/08/2025] [Accepted: 04/29/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND AND OBJECTIVE Our aim was to determine whether induction chemotherapy followed by PARP inhibitor (PARPi) maintenance improves outcomes for patients with metastatic castration-resistant prostate cancer (mCRPC) harboring alterations in homologous recombination repair (HRR) genes in comparison to a historical control cohort treated with PARPi monotherapy. METHODS This single-arm, open-label, investigator-initiated phase 2 trial (NCT02985021) enrolled 18 patients with mCRPC with pathogenic alterations in HRR genes between 2018 and 2021 at a single center. Patients received four cycles of induction chemotherapy with docetaxel (60 mg/m2) and carboplatin (area under the curve 5) every 21 d, followed by maintenance rucaparib (600 mg twice daily) until progression or unacceptable toxicity. The primary outcome was radiographic progression-free survival (rPFS). Subsequent to study inception, multiple other studies reported alterations in genes of the BRCA complex (BRCA-C: BRCA1, BRCA2, PALB2) as most predictive of PARPi response; therefore, a post hoc analysis comparing patients with alterations in BRCA-C genes to a historical control cohort was performed. KEY FINDINGS AND LIMITATIONS After median follow-up of 40.3 mo (interquartile range 38.5-not reached [NR]), the median rPFS for all patients was 8.1 mo (95% confidence interval [CI] 6.5-31.2), similar to a historical control cohort treated with PARPi monotherapy. Among the 12 patients with BRCA-C alterations, median rPFS was 17.7 mo (95% CI 7.5-NR; p = 0.05). A key limitation is the single-arm design. CONCLUSIONS AND CLINICAL IMPLICATIONS Induction platinum-based chemotherapy followed by maintenance PARPi therapy did not improve outcomes for patients with mCRPC broadly selected for HRR deficiency. However, results were promising in the more stringently selected group with BRCA-C gene alterations. Further studies comparing this approach to PARPi monotherapy are warranted.
Collapse
Affiliation(s)
- Ruben Raychaudhuri
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Heather H Cheng
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Roman Gulati
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael T Schweizer
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Aaron Lin
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Todd Yezefski
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hiba M Khan
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Evan Y Yu
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jessica E Hawley
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Peter S Nelson
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Colin C Pritchard
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA
| | - Bruce Montgomery
- Division of Hematology and Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; VA Puget Sound Health Care System, Seattle, WA, USA.
| |
Collapse
|
21
|
Ingrosso G, Lancia A, Bardoscia L, Becherini C, Bottero M, Bertini N, Cai T, Caini S, Caserta C, Doccioli C, Festa E, Francolini G, Giacomelli I, Paolieri F, Scartoni D, Pisani AR, Bellavita R, Livi L, Aristei C, Detti B. Current diagnostic and therapeutic options in de novo low-volume metastatic hormone-sensitive prostate cancer. Expert Rev Anticancer Ther 2025:1-14. [PMID: 40394918 DOI: 10.1080/14737140.2025.2509760] [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: 03/10/2025] [Accepted: 05/19/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION de novo low-volume metastatic hormone-sensitive prostate cancer (mHSPC) patients are characterized by a limited number of metastases at diagnosis. Intensifying the current diagnostic and therapeutic approach including multimodality therapy seems to be key in the clinical management of such patients. AREAS COVERED We comprehensively review the current staging and treatment options for de novo low-volume mHSPC. EXPERT OPINION PSMA-PET should be used in staging high-risk prostate cancer to detect metastatic disease and better stratify patients for individualized treatment. In the era of Androgen Receptor Pathway Inhibitors (ARPIs), Androgen Deprivation Therapy (ADT) alone should be considered an undertreatment for the majority of the patients. Based on current data in the literature, the most effective therapeutic strategy seems to be the combination of intensified systemic treatment (including ADT + ARPI) and radiotherapy for the primary tumor. The role of cytoreductive radical prostatectomy is currently being investigated as well as metastasis-directed therapy to metastatic sites.
Collapse
Affiliation(s)
- Gianluca Ingrosso
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Andrea Lancia
- Department of Radiation Oncology, Policlinico San Matteo Pavia Fondazione IRCCS, Pavia, Italy
| | - Lilia Bardoscia
- Radiation Oncology Unit, S. Luca Hospital, Healthcare Company Tuscany Nord Ovest, Lucca, Italy
| | - Carlotta Becherini
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Marta Bottero
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Niccolò Bertini
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Tommaso Cai
- Department of Urology, Santa Chiara Regional and Teaching Hospital, Trento, Italy
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Saverio Caini
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Claudia Caserta
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Chiara Doccioli
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Eleonora Festa
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Giulio Francolini
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | | | - Federico Paolieri
- Department of Oncology, Hospital of Prato, Azienda USL Toscana Centro, Prato, Italy
| | | | - Antonio Rosario Pisani
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Rita Bellavita
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Lorenzo Livi
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Beatrice Detti
- Radiation Oncology Unit of Prato, Azienda USL Centro Toscana, Prato, Italy
| |
Collapse
|
22
|
Li X, Han Z, Ai J. Synergistic targeting strategies for prostate cancer. Nat Rev Urol 2025:10.1038/s41585-025-01042-6. [PMID: 40394240 DOI: 10.1038/s41585-025-01042-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/22/2025]
Abstract
Prostate cancer is the second most commonly diagnosed cancer and the fifth leading cause of death among men worldwide. Androgen deprivation therapy is a common prostate cancer treatment, but its efficacy is often hindered by the development of resistance, which results in reducing survival benefits. Immunotherapy showed great promise in treating solid tumours; however, clinically significant improvements have not been demonstrated for patients with prostate cancer, highlighting specific drawbacks of this therapeutic modality. Hence, exploring novel strategies to synergistically enhance the efficacy of prostate cancer immunotherapy is imperative. Clinical investigations have focused on the combined use of targeted or gene therapy and immunotherapy for prostate cancer. Notably, tumour-specific antigens and inflammatory mediators are released from tumour cells after targeted or gene therapy, and the recruitment and infiltration of immune cells, including CD8+ T cells and natural killer cells activated by immunotherapy, are further augmented, markedly improving the efficacy and prognosis of prostate cancer. Thus, immunotherapy, targeted therapy and gene therapy could have reciprocal synergistic effects in prostate cancer in combination, resulting in a proposed synergistic model encompassing these three therapeutic modalities, presenting novel potential treatment strategies for prostate cancer.
Collapse
Affiliation(s)
- Xuanji Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zeyu Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Vescovo M, Raspollini MR, Nibid L, Castiglione F, Nardi E, de Biase D, Massari F, Giunchi F, Pepe F, Troncone G, Malapelle U, Carosi M, Casini B, Melucci E, Fassan M, Toffolatti L, Guerini-Rocco E, Conversano F, Rappa A, Tommasi S, Coppola CA, Zeppa P, Caputo A, Gaeta S, Pagni F, Seminati D, Vecchione A, Scarpino S, Righi D, Taffon C, Prata F, Perrone G. Storage Time and DNA Quality Determine BRCA1/2 Sequencing Success in Prostate Cancer: A Multicentre Analysis with Therapeutic Implications. Cancers (Basel) 2025; 17:1705. [PMID: 40427202 PMCID: PMC12110138 DOI: 10.3390/cancers17101705] [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: 04/08/2025] [Revised: 05/04/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Approximately 25.0% of metastatic prostate cancer patients harbour DNA damage repair mutations, including BRCA1 and BRCA2, which are actionable targets for poly(ADP-ribose) polymerase (PARP) inhibitors. Accurate detection of BRCA1/2 mutations is critical for guiding targeted therapies, but crucial pre-analytical factors, such as tissue storage duration and DNA fragmentation, drastically affect the reliability of next-generation sequencing (NGS) using real-world diagnostic specimens. METHODS This multicentre study analysed 954 formalin-fixed paraffin-embedded tissue samples from 11 centres, including 559 biopsies and 395 surgical specimens. This study examined the impact of storage duration (<1 year, 1-2 years, and >2 years) and DNA parameters (concentration and fragmentation index) on NGS success rates. Logistic regression and Cox regression analyses were used to assess correlations between these factors and sequencing outcomes. RESULTS NGS success rates decreased significantly with longer storage, from 87.8% (<1 year) to 69.1% (>2 years). Samples with higher DNA concentrations and fragmentation indexes had higher success rates (p < 0.001). Surgical specimens had superior success rates (83.3%) compared with biopsies (72.8%) due to better DNA quality. The DNA degradation rate was more pronounced in older samples, underscoring the negative impact of extended storage. CONCLUSIONS Timely testing of BRCA1/2 mutations is critical for optimizing the identification of prostate cancer patients eligible for PARP inhibitors. Surgical specimens provide more reliable results than biopsies and minimizing the storage duration significantly enhances testing outcomes. Standardizing pre-analytical and laboratory procedures across centres is essential to ensure personalized treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Mariavittoria Vescovo
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
| | - Maria Rosaria Raspollini
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Via Pieraccini, 6, 50129 Florence, Italy; (M.R.R.); (F.C.); (E.N.)
| | - Lorenzo Nibid
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Francesca Castiglione
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Via Pieraccini, 6, 50129 Florence, Italy; (M.R.R.); (F.C.); (E.N.)
| | - Eleonora Nardi
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Via Pieraccini, 6, 50129 Florence, Italy; (M.R.R.); (F.C.); (E.N.)
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (F.P.); (G.T.); (U.M.)
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (F.P.); (G.T.); (U.M.)
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (F.P.); (G.T.); (U.M.)
| | - Mariantonia Carosi
- Molecular Diagnostic Laboratory, Pathology Department, Advanced Diagnostics Research and Technological Innovation Department, Regina, Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (M.C.); (B.C.); (E.M.)
| | - Beatrice Casini
- Molecular Diagnostic Laboratory, Pathology Department, Advanced Diagnostics Research and Technological Innovation Department, Regina, Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (M.C.); (B.C.); (E.M.)
| | - Elisa Melucci
- Molecular Diagnostic Laboratory, Pathology Department, Advanced Diagnostics Research and Technological Innovation Department, Regina, Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (M.C.); (B.C.); (E.M.)
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy;
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy
| | - Luisa Toffolatti
- Surgical Pathology Unit, Ca’ Foncello General Hospital, ULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Elena Guerini-Rocco
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy;
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (F.C.); (A.R.)
| | - Federica Conversano
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (F.C.); (A.R.)
| | - Alessandra Rappa
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (F.C.); (A.R.)
| | - Stefania Tommasi
- Pharmacogenetics and Molecular Diagnostics Unit, IRCCS Istituto Tumori Giovanni Paolo II Bari, 70124 Bari, Italy; (S.T.); (C.A.C.)
| | - Claudio Antonio Coppola
- Pharmacogenetics and Molecular Diagnostics Unit, IRCCS Istituto Tumori Giovanni Paolo II Bari, 70124 Bari, Italy; (S.T.); (C.A.C.)
| | - Pio Zeppa
- Department of Medicine, Surgery, and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84084 Baronissi, Italy; (P.Z.); (A.C.)
| | - Alessandro Caputo
- Department of Medicine, Surgery, and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84084 Baronissi, Italy; (P.Z.); (A.C.)
| | - Sara Gaeta
- Department of Pathology, University Hospital of Salerno, 84081 Salerno, Italy;
| | - Fabio Pagni
- Department of Medicine and Surgery, University Milan Bicocca and Fondazione IRCCS San Gerardo dei Tintori Monza, 20126 Milan, Italy; (F.P.)
| | - Davide Seminati
- Department of Medicine and Surgery, University Milan Bicocca and Fondazione IRCCS San Gerardo dei Tintori Monza, 20126 Milan, Italy; (F.P.)
| | - Andrea Vecchione
- Unit of Pathology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (A.V.); (S.S.)
| | - Stefania Scarpino
- Unit of Pathology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (A.V.); (S.S.)
| | - Daniela Righi
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
| | - Chiara Taffon
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Francesco Prata
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
| | - Giuseppe Perrone
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| |
Collapse
|
24
|
Fang W, Chen Y, Nie M, Zhou X, Liu Y, Tao H, Yang B, Wang X. Targeting YY1-DR5 Axis by Pyripyropene O as a Novel Therapeutic Strategy Against Prostate Cancer: Molecular Mechanisms and In Vivo Zebrafish Validation. Mar Drugs 2025; 23:214. [PMID: 40422804 DOI: 10.3390/md23050214] [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: 04/15/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Induction of apoptosis is an important strategy for the treatment of prostate cancer. DR5 is a member of the death receptor superfamily and targeting DR5 is an effective way to induce apoptosis. Pyripyropene O is a natural compound isolated from the marine fungus Aspergillus fumigatus SCSIO 41220. We found it has anti-prostate cancer potential by inducing apoptosis; Methods: The effects of pyripyropene O on the viability, proliferation, cell cycle, apoptosis and migration of prostate cancer cells were investigated by MTT assay, plate clone formation assay, 3D cell sphere assay, flow cytometry and real-time cell analysis. Transmission electron microscopy was used to observe the changes in the internal structure of prostate cancer cells after treatment with pyripyropene O. After determining the mode of cell death, the mechanism of action of pyripyropene O on prostate cancer was further investigated using apoptotic protein microarray, western blot, qPCR, molecular docking, cellular immunofluorescence staining and cellular thermal shift assay. After explaining the mechanism of action of pyriproxyfen O, the in vivo absorption, distribution, metabolism, excretion and potential toxicity of pyriproxyfen O were investigated using ADMETLab 2.0 software. Finally, a zebrafish xenograft tumour model was developed to evaluate the anti-prostate cancer effects of pyriproxyfen O in vivo; Results: The experimental results at the cellular level showed that pyripyropene O inhibited the survival, proliferation and migration of prostate cancer cells, and also showed that pyripyropene O blocked the prostate cancer cell cycle at the G2/M phase and induced apoptosis. At the molecular level, pyripyropene O binds to the transcription factor YY1, promotes YY1 nuclear translocation, regulates the transcription level of DR5, a target gene of YY1, and upregulates the expression of DR5 mRNA and protein. The in vivo results showed that pyripyropene O effectively inhibited the development of prostate cancer in zebrafish; Conclusions: Pyripyropene O has a clear anti-prostate cancer effect at both cellular and animal levels, inhibiting the survival and proliferation of prostate cancer cells by binding to the transcription factor YY1 to activate the expression of DR5 to promote apoptosis, thus exerting an inhibitory effect on prostate cancer.
Collapse
Affiliation(s)
- Wenxuan Fang
- Guangxi Engineering Research Center for High-Value Utilization of Guangxi-Produced Authentic Medicinal Herbs, Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Ying Chen
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Mingyi Nie
- Guangxi Engineering Research Center for High-Value Utilization of Guangxi-Produced Authentic Medicinal Herbs, Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xuefeng Zhou
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Yonghong Liu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Huaming Tao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Bin Yang
- Guangdong Key Laboratory of Marine Materia Medica/State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Xueni Wang
- Guangxi Engineering Research Center for High-Value Utilization of Guangxi-Produced Authentic Medicinal Herbs, Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
25
|
Smith SC, Melson JW, Quillin JM, Hiemenz MC, Tomlins SA, Wobker SE. A pathologist's primer on implementing new standard-of-care molecular biomarker testing for precision prostate cancer management. Am J Clin Pathol 2025; 163:649-655. [PMID: 39838622 DOI: 10.1093/ajcp/aqae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025] Open
Affiliation(s)
- Steven C Smith
- Departments of Pathology and Surgery, VCU School of Medicine, VCU Massey Comprehensive Cancer Center, and Richmond VA Medical Center, Richmond, VA, United States
| | - John W Melson
- Department of Medicine, VCU School of Medicine and VCU Massey Comprehensive Cancer Center, Richmond, VA, United States
| | - John M Quillin
- Department of Pediatrics, VCU School of Medicine, Richmond, VA, United States
| | | | - Scott A Tomlins
- Strata Oncology and Departments of Pathology and Urology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Sara E Wobker
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
26
|
Elshimy Y, Alkhatib AR, Atassi B, Mohammad KS. Biomarker-Driven Approaches to Bone Metastases: From Molecular Mechanisms to Clinical Applications. Biomedicines 2025; 13:1160. [PMID: 40426987 PMCID: PMC12109438 DOI: 10.3390/biomedicines13051160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Bone metastases represent a critical complication in oncology, frequently indicating advanced malignancy and substantially reducing patient quality of life. This review provides a comprehensive analysis of the complex interactions between tumor cells and the bone microenvironment, emphasizing the relevance of the "seed and soil" hypothesis, the RANK/RANKL/OPG signaling axis, and Wnt signaling pathways that collectively drive metastatic progression. The molecular and cellular mechanisms underlying the formation of osteolytic and osteoblastic lesions are examined in detail, with a particular focus on their implications for bone metastases associated with breast, prostate, lung, and other cancers. A central component of this review is the categorization of pathological biomarkers into four types: diagnostic, prognostic, predictive, and monitoring. We provide a comprehensive evaluation of circulating tumor cells (CTCs), bone turnover markers (such as TRACP-5b and CTX), advanced imaging biomarkers (including PET/CT and MRI), and novel genomic signatures. These biomarkers offer valuable insights for early detection, enhanced risk stratification, and optimized therapeutic decision-making. Furthermore, emerging strategies in immunotherapy and bone-targeted treatments are discussed, highlighting the potential of biomarker-guided precision medicine to enhance personalized patient care. The distinctiveness of this review lies in its integrative approach, combining fundamental pathophysiological insights with the latest developments in biomarker discovery and therapeutic innovation. By synthesizing evidence across various cancer types and biomarker categories, we provide a cohesive framework aimed at advancing both the scientific understanding and clinical management of bone metastases.
Collapse
Affiliation(s)
| | | | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (Y.E.); (A.R.A.); (B.A.)
| |
Collapse
|
27
|
Szalontai J, Szarvas T, Miszczyk M, Nyirády P, Shariat SF, Fazekas T. Toxicities of PARP inhibitors in genitourinary cancers. Curr Opin Urol 2025:00042307-990000000-00251. [PMID: 40336260 DOI: 10.1097/mou.0000000000001297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
PURPOSE OF REVIEW Recent advancements in the understanding of the genetic background of genitourinary cancers allowed for a successful introduction of targeted antitumor agents to prostate cancer (PCa) treatment. Inhibitors of the poly ADP-ribose polymerase enzyme (PARPi) transformed the treatment landscape of metastatic prostate cancer, and being increasingly studied in earlier disease stages. However, they are associated with nonnegligible toxicity, therefore, we aimed to summarize their side-effect profile in patients with PCa. RECENT FINDINGS Hematologic toxicities, particularly anemia, thrombocytopenia, and neutropenia are among the most common and serious adverse events associated with PARPi, highlighting the need for regular blood count monitoring. Nonhematologic side effects, including fatigue, nausea, vomiting, diarrhea, and constipation, are common, and can be mitigated with supportive interventions like dietary modifications, antiemetics, or stool management techniques. Special attention should be given to patients with therapy-resistant or persistent cytopenia, in whom bone marrow biopsy should be considered, as it can indicate myelodysplastic syndrome and acute myeloid leukemia. SUMMARY PARP inhibitors represent a major advancement in the management of metastatic prostate cancer, offering a significant survival benefit in applicable cases. However, patients need to be carefully selected and informed, to allow for optimal balancing between the benefits and nonneglectable risks of severe toxicities. Better understanding of PARPi toxicity profile can improve personalized decision-making and enhance treatment compliance, through raising patients' awareness about the possible side effects of PARPi.
Collapse
Affiliation(s)
- János Szalontai
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Tibor Szarvas
- Department of Urology, Semmelweis University, Budapest, Hungary
- Department of Urology, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Marcin Miszczyk
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Collegium Medicum - Faculty of Medicine, WSB University, Dąbrowa Górnicza, Poland
| | - Péter Nyirády
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Shahrokh F Shariat
- Department of Urology, Semmelweis University, Budapest, Hungary
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Urology, Weill Cornell Medical College, New York, New York, USA
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
- Research Centre for Evidence Medicine, Urology Department, Tabriz University of Medical Sciences, Tabriz, Iran
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| | - Tamás Fazekas
- Department of Urology, Semmelweis University, Budapest, Hungary
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Simoneau A, Pratt CB, Wu HJ, Rajeswaran SS, Comer CG, Sudsakorn S, Zhang W, Liu S, Meier SR, Choi AH, Khendu T, Stowe H, Shen B, Whittington DA, Chen Y, Yu Y, Mallender WD, Feng T, Andersen JN, Maxwell JP, Throner S. Characterization of TNG348: A Selective, Allosteric USP1 Inhibitor That Synergizes with PARP Inhibitors in Tumors with Homologous Recombination Deficiency. Mol Cancer Ther 2025; 24:678-691. [PMID: 39886906 PMCID: PMC12046316 DOI: 10.1158/1535-7163.mct-24-0515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/01/2025]
Abstract
Inhibition of the deubiquitinating enzyme USP1 can induce synthetic lethality in tumors characterized by homologous recombination deficiency (HRD) and represents a novel therapeutic strategy for the treatment of BRCA1/2-mutant cancers, potentially including patients whose tumors have primary or acquired resistance to PARP inhibitors (PARPi). In this study, we present a comprehensive characterization of TNG348, an allosteric, selective, and reversible inhibitor of USP1. TNG348 induces dose-dependent accumulation of ubiquitinated protein substrates both in vitro and in vivo. CRISPR screens show that TNG348 exerts its antitumor effect by disrupting the translesion synthesis pathway of DNA damage tolerance through RAD18-dependent ubiquitinated PCNA. Although TNG348 and PARPi share the ability to selectively kill HRD tumor cells, CRISPR screens reveal that TNG348 and PARPi do so through discrete mechanisms. Particularly, knocking out PARP1 causes resistance to PARPi but sensitizes cells to TNG348 treatment. Consistent with these findings, combination of TNG348 with PARPi leads to synergistic antitumor effects in HRD tumors, resulting in tumor growth inhibition and regression in multiple mouse xenograft tumor models. Importantly, our data on human cancer models further show that the addition of TNG348 to PARPi treatment can overcome acquired PARPi resistance in vivo. Although the clinical development of TNG348 has been discontinued because of unexpected liver toxicity in patients (NCT06065059), the present data provide preclinical and mechanistic support for the continued exploration of USP1 as a drug target for the treatment of patients with BRCA1/2-mutant or HRD cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yi Yu
- Tango Therapeutics, Boston, Massachusetts
| | | | | | | | | | | |
Collapse
|
29
|
Garje R, Riaz IB, Naqvi SAA, Rumble RB, Taplin ME, Kungel TM, Herchenhorn D, Zhang T, Beckermann KE, Vapiwala N, Carducci MA, Celano P, Hotte SJ, Basu A, Borno H, Bryce AH, Wang P, Wulff-Burchfield E, Bodei L, Loblaw A, Hamilton RJ, Emamekhoo H, Hope TA, He H, Murad MH, Liu H, Williams JE, Parikh RA. Systemic Therapy in Patients With Metastatic Castration-Resistant Prostate Cancer: ASCO Guideline Update. J Clin Oncol 2025:JCO2500007. [PMID: 40315400 DOI: 10.1200/jco-25-00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/13/2025] [Indexed: 05/04/2025] Open
Abstract
PURPOSE To provide evidence-based recommendations for patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS An Expert Panel including patient representation completed a systematic review of the evidence and made recommendations. RESULTS Depending upon prior treatment received, androgen receptor pathway inhibitors (ARPIs: enzalutamide, abiraterone with prednisone), poly(ADP-ribose) polymerase inhibitors (PARPi), chemotherapeutic agents (docetaxel, cabazitaxel), radiopharmaceuticals (radium 223, 177Lu-prostate-specific membrane antigen [PSMA]-617), and sipuleucel-T have demonstrated an overall survival (OS) benefit for patients with mCRPC. For patients with BRCA1/2 alterations who did not receive prior ARPI, the combination of PARPi and ARPI (talazoparib + enzalutamide, olaparib and/or niraparib + abiraterone) has shown clinical benefit. For patients with BRCA1/2 alterations who received prior ARPI or ARPI followed by docetaxel, olaparib showed OS benefit. In select patients with microsatellite instability-high/mismatch repair-deficient, pembrolizumab showed clinical efficacy. RECOMMENDATIONS Prior systemic therapy for castration-sensitive prostate cancer will determine subsequent therapy used for mCRPC. Continue androgen-deprivation therapy for patients with mCRPC indefinitely. Early adoption of somatic genetic testing and palliative care is recommended. Patients with mCRPC and bony metastases should receive a bone-protective agent. The panel recommends the combination of ARPI with PARPi in patients with BRCA1/2 alterations who did not receive prior ARPI. For patients who received prior ARPI, the panel recommends docetaxel chemotherapy. The panel recommends 177Lu-PSMA-617 or cabazitaxel chemotherapy for patients who receive prior ARPI and docetaxel chemotherapy. For patients with BRCA1/2 alterations who received prior ARPI, the panel recommends PARPi monotherapy. Radium 223 is recommended for patients with symptomatic bone-only disease. Evidence for optimal sequencing for mCRPC regimens is lacking.Additional information is available at www.asco.org/genitourinary-cancer-guidelines.
Collapse
Affiliation(s)
- Rohan Garje
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | | | | | | | | | | | - Daniel Herchenhorn
- Instituto D'Or/Oncologia D'Or, Latin America Cooperative Group (LACOG)- Genito-Urinary, Rio de Janeiro, Brazil
| | - Tian Zhang
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | | | - Neha Vapiwala
- University of Pennsylvania Abramson Cancer Center, Philadelphia, PA
| | | | - Paul Celano
- Greater Baltimore Medical Center (GBMC), Towson, MD
| | | | - Arnab Basu
- University of Alabama at Birmingham, Birmingham, AL
| | - Hala Borno
- University of California, San Francisco, San Francisco, CA
| | | | - Peng Wang
- Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Lisa Bodei
- Weill Cornell Medical College of Cornell University, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew Loblaw
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Robert J Hamilton
- Cancer Clinical Research Unit, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Thomas A Hope
- University of California, San Francisco, San Francisco, CA
| | - Huan He
- Yale University, New Haven, CT
| | | | | | | | | |
Collapse
|
30
|
Kamii M, Kamata R, Saito H, Yamamoto G, Mashima C, Yamauchi T, Nakao T, Sakae Y, Yamamori-Morita T, Nakai K, Hakozaki Y, Takenaka M, Okamoto A, Ohashi A. PARP inhibitors elicit a cellular senescence mediated inflammatory response in homologous recombination proficient cancer cells. Sci Rep 2025; 15:15458. [PMID: 40316566 PMCID: PMC12048520 DOI: 10.1038/s41598-025-00336-4] [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: 01/10/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have improved the prognosis of homologous recombination deficient (HRD) ovarian cancer (OC), while effective therapeutic strategies for HR-proficient (HRP) OC still need to be established. This study investigates senescence-mediated inflammation as a novel mechanism of action for PARP inhibitors in HRP cancers. Transcriptome analyses were performed in olaparib-treated HeLa cells as a HRP model. Interferon regulatory factor-Lucia luciferase (IRF-Luc) reporter activity was assessed. The effects of PARP inhibitors on senescence-like phenotypes were assessed in seven HRP cancer cell lines, based on morphological changes, senescence-associated β-galactosidase (SA-β-GAL) activity, cellular granularity, and senescence-associated secretory phenotype (SASP)-related gene expression. Peripheral blood mononuclear cell (PBMC) migration assays were also performed with the conditioned medium in treatment with the PARP inhibitor. Transcriptome analyses revealed numbers of inflammatory cytokine- and chemokine-related pathways were significantly upregulated in olaparib-treated HeLa cells, which were confirmed by IRF-Luc reporter assays. The PARP inhibitors induced senescent phenotypes in HRP cancer cell lines: flattened and enlarged morphology, increased SA-β-GAL activity, elevated cellular granularity, and upregulated expressions of SASP-related genes (e.g., IL1B, IL6, and CXCL10). Furthermore, in vitro migration assays revealed that PARP inhibitor-treated HRP cancer cells attracted PBMCs more abundantly, suggesting the potential for recruiting immune cells to HRP cancer cells through senescence-mediated immunological activation. Our findings suggest that PARP inhibitors recruit immune cells to HRP cancer cells, potentially activating immune responses in the tumor microenvironment, providing new insights into the clinical benefits of PARP inhibitors in immunotherapy for patients with HRP OC.
Collapse
Affiliation(s)
- Misato Kamii
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ryo Kamata
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Hitoshi Saito
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Gaku Yamamoto
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Chiaki Mashima
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Toyohiro Yamauchi
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Department of Integrated Bioscience, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan
| | - Takehiro Nakao
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yuta Sakae
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Tomoko Yamamori-Morita
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Kazuki Nakai
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yumi Hakozaki
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Masataka Takenaka
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Akihiro Ohashi
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
- Department of Integrated Bioscience, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan.
| |
Collapse
|
31
|
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] [Download PDF] [Figures] [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
|
32
|
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
|
33
|
Dang M, Schritz A, Goncharenko N, Berchem G. Impact of molecular diagnostics and targeted cancer therapy on patient outcomes (MODIFY): a retrospective study of the implementation of precision oncology. Mol Oncol 2025; 19:1508-1516. [PMID: 39661533 PMCID: PMC12077269 DOI: 10.1002/1878-0261.13785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/08/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
High-throughput genomic analyses are being implemented in clinical practice. MODIFY is a retrospective study of the first introduction of genomic profiling and molecular tumor boards in the country of Luxembourg. The primary objective was to assess whether patients derived a clinical benefit by measuring the percentage of patients who presented a progression-free survival (PFS) on matched therapy (PFS2) 1.3-fold longer than PFS on previous therapy (PFS1). A total of 94 patients were included. In total, 45 patients (53.57% of patients with successful next-generation sequencing [NGS] analysis) were found to have an actionable mutation. Of these, 11 patients received the treatment recommended by the molecular tumor board, another 12 received best-supportive care, and 20 were treated with conventional therapy. PFS2 and PFS1 data were available for eight patients. The PFS2/PFS1 ratio was ≥ -1.3 in 62.5% (n = 5/8; CI [30.38, 86.51]) of patients; three patients showed a partial response, and median overall survival (OS) was 7.3 months. Although the examined population was small, this study further supports evidence indicating that patients with advanced cancer benefit from molecular profiling and targeted therapy.
Collapse
Affiliation(s)
- Michaël Dang
- Department of OncologyCentre Hospitalier de Luxembourg (CHL)Luxembourg
- University of Luxembourg (Uni.lu)Luxembourg
| | | | | | - Guy Berchem
- Department of OncologyCentre Hospitalier de Luxembourg (CHL)Luxembourg
- University of Luxembourg (Uni.lu)Luxembourg
- Institut National du Cancer (INC)LuxembourgLuxembourg
| |
Collapse
|
34
|
Fu J, Franzen N, Aas E, Koen van der Mijn JC, van Leeuwen PJ, Retel VP. Early Cost-Effectiveness Analysis of Using Whole-Genome Sequencing for Patients With Castration-Resistant Prostate Cancer. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:720-729. [PMID: 40049327 DOI: 10.1016/j.jval.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/16/2025] [Accepted: 02/16/2025] [Indexed: 03/30/2025]
Abstract
OBJECTIVES This study aims to assess the potential cost-effectiveness of using whole-genome sequencing (WGS)-guided systemic therapy in metastatic castrate-resistant prostate cancer compared with the European Association of Urology guideline recommended diagnostics from a Dutch societal perspective. METHODS A decision analytic model combining a decision tree and partitioned survival models was developed to link diagnostic results with subsequent biomarker-guided treatments. Two diagnostic strategies, WGS and guideline-recommended practice-the genomic testing for breast cancer gene 1/2 (BRCA1/2) and deficient mismatch repair, were simulated to compare the health outcome and cost. Treatment effectiveness was estimated through survival analysis using published trial data. Sensitivity and scenario analyses were conducted to examine result robustness and to identify conditions under which WGS may be cost-effective. RESULTS WGS identified an additional 21% of patients eligible for personalized therapy (PD-1/PDL-1 inhibitors and olaparib), resulting in an incremental increase in cost (€14 260) and quality-adjusted life years (QALY = 0.05). These results yielded an incremental cost-effectiveness ratio of €289 625 per QALY gained. WGS would become cost-effective if the cost of biomarker-guided therapies decreases by 62% and when identifying a proportion of 23% more patients with actional targets. CONCLUSIONS Our findings suggest that future treatments with improved efficacy and reduced cost could potentially make the WGS strategy cost-effective. Its unaccounted potential value to identify prognostic biomarkers, diagnostic alternatives, and patient heterogeneity should be addressed in future research and considered for optimal implementation. New reimbursement options are needed considering the high prices of biomarker-guided therapies that drive the incremental cost-effectiveness ratio.
Collapse
Affiliation(s)
- Jinjing Fu
- Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Nora Franzen
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Eline Aas
- Department of Health Management and Health Economics, University of Oslo, Oslo, Norway; Division of Health Services, Norwegian Institute of Public Health, Oslo, Norway
| | - J C Koen van der Mijn
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Pim J van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Valesca P Retel
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands; Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
Turco F, Gillessen S, Herrmann K, Paone G, Omlin A. Treatment Landscape of Prostate Cancer in the Era of PSMA Radiopharmaceutical Therapy. J Nucl Med 2025; 66:665-672. [PMID: 40015917 DOI: 10.2967/jnumed.124.267730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/29/2025] [Indexed: 03/01/2025] Open
Abstract
The treatment landscape of prostate cancer is quite complex because of the many therapeutic options available in different disease settings (hormonal treatments, chemotherapy, poly(adenosine diphosphate ribose) polymerase inhibitors, radiopharmaceutical therapy). Since in most cases we do not have comparative studies between these different agents, the best therapeutic sequence in patients with prostate cancer remains unsolved. In this review, we describe the different systemic therapeutic options available in each disease setting from localized disease to metastatic castration-resistant disease. We also indicate when to use each of these therapeutic options in the therapeutic sequence on the basis of the results of the available studies. A special focus of this review is the place of prostate-specific membrane antigen radiopharmaceutical therapy in the treatment algorithms.
Collapse
Affiliation(s)
- Fabio Turco
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland;
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium, University Hospital Essen, Essen, Germany
| | - Gaetano Paone
- Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland
- Clinic of Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; and
| | - Aurelius Omlin
- Onkozentrum Zurich, University of Zurich and Tumorzentrum Hirslanden Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Habaka M, Daly GR, Shinyanbola D, Alabdulrahman M, McGrath J, Dowling GP, Hehir C, Huang HYR, Hill ADK, Varešlija D, Young LS. PARP Inhibitors in the Neoadjuvant Setting; A Comprehensive Overview of the Rationale for their Use, Past and Ongoing Clinical Trials. Curr Oncol Rep 2025; 27:533-551. [PMID: 40192976 DOI: 10.1007/s11912-025-01669-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 05/16/2025]
Abstract
PURPOSEOF REVIEW Poly (ADP-ribose) polymerases (PARPs) are enzymes essential for detecting and repairing DNA damage through poly-ADP-ribosylation. In cancer, cells with deficiencies in homologous recombination repair mechanisms often become more dependent on PARP-mediated repair mechanisms to effectively repair dsDNA breaks. As such, PARP inhibitors (PARPis) were introduced into clinical practice, serving as a key targeted therapy option through synthetic lethality in the treatment of cancers with homologous recombination repair deficiency (HRD). Though PARPis are currently approved in the adjuvant setting for several cancer types such as ovarian, breast, prostate and pancreatic cancer, their potential role in the neoadjuvant setting remains under investigation. This review outlines the rationale for using PARPi in the neoadjuvant setting and evaluates findings from early and ongoing clinical trials. RECENT FINDINGS Our analysis indicates that numerous studies have explored PARPi as a neoadjuvant treatment for HRD-related cancers. The majority of neoadjuvant PARPi trials have been performed in breast and ovarian cancer, while phase II/III evidence supporting efficacy in prostate and pancreatic cancers remains limited. Studies are investigating PARPi in the neoadjuvant setting of HRD-related cancers. Future research should prioritize combination strategies with immune checkpoint inhibitors and expand outcome measures to include patient satisfaction and quality-of-life metrics.
Collapse
Affiliation(s)
- Minatoullah Habaka
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - Gordon R Daly
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Deborah Shinyanbola
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Jason McGrath
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gavin P Dowling
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Cian Hehir
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Helen Ye Rim Huang
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Arnold D K Hill
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| | - Damir Varešlija
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| | - Leonie S Young
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
37
|
Montaldo NP, Nilsen HL, Bordin DL. Targeting base excision repair in precision oncology. DNA Repair (Amst) 2025; 149:103844. [PMID: 40359788 DOI: 10.1016/j.dnarep.2025.103844] [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: 01/31/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
Targeting the DNA damage response (DDR) is a key strategy in cancer therapy, leveraging tumour-specific weaknesses in DNA repair pathways to enhance treatment efficacy. Traditional treatments, such as chemotherapy and radiation, use a broad, damage-inducing approach, whereas precision oncology aims to tailor therapies to specific genetic mutations or vulnerabilities. The clinical success of PARP inhibitors has renewed the interest in targeting DNA repair as a therapeutic strategy. Expanding the precision oncology toolbox by targeting the base excision repair (BER) pathway presents a promising avenue for cancer therapy, particularly in tumours that rely heavily on this pathway due to deficiencies in other DNA repair mechanisms. This review discusses how targeting BER could improve treatment outcomes, particularly in DDR-defective cancers. With ongoing advancements in biomarker discovery and drug development, BER-targeted therapies hold significant potential for refining precision oncology approaches.
Collapse
Affiliation(s)
- Nicola P Montaldo
- Department of Microbiology, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway; CRESCO - Centre for embryology and healthy Development, University of Oslo, Norway
| | - Hilde Loge Nilsen
- Department of Microbiology, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway; CRESCO - Centre for embryology and healthy Development, University of Oslo, Norway.
| | - Diana L Bordin
- Akershus University Hospital, Department of Clinical Molecular Biology, Unit for Precision Medicine, Lørenskog, Norway
| |
Collapse
|
38
|
Bitting RL, McNair C, Wyatt AW, Vandekerkhove G, Choi T, Leader AE, Blanding-Godbolt J, Gross L, Hamade K, Halabi S, Giri VN. Factors Affecting Genomic Testing in Prostate Cancer: Results From the Decision-Making, Experience, and Confidence In Determining Genomic Evaluation (DECIDE) Survey. JCO Precis Oncol 2025; 9:e2400821. [PMID: 40373262 DOI: 10.1200/po-24-00821] [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: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/28/2025] [Indexed: 05/17/2025] Open
Abstract
PURPOSE Genomic testing for prostate cancer (PCa) clinical management and hereditary cancer assessment has grown in clinical impact; however, challenges remain regarding optimal implementation and end-user confidence. The Decision-making, Experience, and Confidence In Determining Genomic Evaluation (DECIDE) survey was designed to collect information regarding utility and understanding of genomic testing from PCa health care providers, researchers, and stakeholders. METHODS The DECIDE survey was administered online from October 2022 to January 2023 with 18 multiple-response questions. Survey domains included self-confidence with ordering and interpreting germline and somatic genomic tests, process of testing and use of results, decision-making factors, and barriers to testing. Data were summarized by evaluating counts and percentages of responses, and the results were presented by descriptive statistics. RESULTS One hundred twenty-two participants completed the survey. The majority were medical oncologists (70%) and at academic medical centers (89%). Self-confidence was high in knowing indications for genomic testing (82% respondents) but lower in interpretation of results, especially from circulating tumor DNA (52%). Confidence varied in interpreting pathogenic variants (65% high confidence), variants of unknown significance (47%), and incidental findings from genomic tests (35%). Common barriers to testing were difficulty obtaining tissue (71%) and cost (35%). Testing utility was sometimes limited by inability to obtain the recommended treatment (33%). Most of the respondents (55%) agreed that lack of education and training of health care professionals regarding genomic testing is impeding clinical translation. CONCLUSION The DECIDE survey provided critical insights into challenges with genomic testing, from provider confidence in interpretating results to testing and practice barriers. The results inform next steps to further educate PCa providers and to collectively improve testing and result reporting for enhanced implementation of PCa genomic testing.
Collapse
Affiliation(s)
- Rhonda L Bitting
- Duke Cancer Institute, Duke University, Durham, NC
- Duke University School of Medicine, Durham, NC
- Durham VA Healthcare System, Durham, NC
| | - Christopher McNair
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Alexander W Wyatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
- Michael Smith Genome Sciences Centre and Clinical Cancer Genomics Program, BC Cancer, Vancouver, Canada
| | | | - Taehwa Choi
- School of Mathematics, Statistics and Data Science, Sungshin Women's University, Seoul, South Korea
| | - Amy E Leader
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | - Laura Gross
- Yale University School of Medicine, Yale Cancer Center, New Haven, CT
| | - Khaldoun Hamade
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Susan Halabi
- Duke Cancer Institute, Duke University, Durham, NC
- Duke University School of Medicine, Durham, NC
| | - Veda N Giri
- Yale University School of Medicine, Yale Cancer Center, New Haven, CT
| |
Collapse
|
39
|
Saad F, Armstrong AJ, Shore N, George DJ, Oya M, Sugimoto M, McKay RR, Hussain M, Clarke NW. Olaparib Monotherapy or in Combination with Abiraterone for the Treatment of Patients with Metastatic Castration-Resistant Prostate Cancer (mCRPC) and a BRCA Mutation. Target Oncol 2025; 20:445-466. [PMID: 40397306 DOI: 10.1007/s11523-025-01146-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2025] [Indexed: 05/22/2025]
Abstract
Treatment strategies to improve outcomes in patients with metastatic castration-resistant prostate cancer (mCRPC) are evolving. Of particular interest are therapies that target DNA damage responses in tumor cells by inhibiting poly(ADP-ribose) polymerase (PARP) activity. Several PARP inhibitors have recently received regulatory approval for the treatment of patients with mCRPC, of which olaparib was the first for prostate cancer. Olaparib received approval as a monotherapy following the PROfound study (NCT02987543) and in combination with abiraterone following the PROpel study (NCT03732820) for mCRPC. Both PROfound (homologous recombination repair mutation biomarker-selected) and PROpel (biomarker unselected) patients demonstrated statistically significant longer radiographic progression-free survival (rPFS) with olaparib versus their respective control arms in the intention-to-treat population. In both studies, the greatest clinical benefit with olaparib was seen in patients with BRCA1 and/or BRCA2 mutations (BRCAm): PROfound rPFS hazard ratio (HR) 0.22 (95% confidence interval [CI] 0.15-0.32); PROpel rPFS HR 0.23 (95% CI 0.12-0.43). Clinical benefit was also observed in terms of overall survival: PROfound HR 0.63 (95% CI 0.42-0.95); PROpel HR 0.29 (95% CI 0.14-0.56). We provide a comprehensive overview of the utility of olaparib for patients with mCRPC harboring a BRCAm. Key clinical and safety data in BRCAm subgroup populations are discussed, predominantly based on findings from PROfound and PROpel, as well as investigator-initiated studies, to help inform treatment decision-making in this patient population. We also discuss the importance of genetic testing to identify patients who may optimally benefit from treatment with olaparib, either as a monotherapy or in combination with abiraterone.
Collapse
Affiliation(s)
- Fred Saad
- Division of Urology and GU Oncology, Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, QC, Canada.
| | - Andrew J Armstrong
- Divisions of Medical Oncology and Urology, Duke Cancer Institute Center for Prostate and Urologic Cancer, Duke University, Durham, NC, USA
| | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Daniel J George
- Department of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mikio Sugimoto
- Department of Urology, Kagawa University Hospital, Kagawa, Japan
| | - Rana R McKay
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Maha Hussain
- Division of Hem/Onc, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Noel W Clarke
- Department of Surgery, The Christie and Salford Royal NHS Foundation Trusts, Manchester, UK
| |
Collapse
|
40
|
Kim KB, Desprez PY, de Semir D, Woo RWL, Sharma A, Jones R, Caressi C, Nosrati M, Janiczek E, Rivera Penafiel J, Kashani-Sabet M. Phase II Study of Niraparib in Patients With Advanced Melanoma With Homologous Recombination Pathway Gene Mutations. JCO Precis Oncol 2025; 9:e2400658. [PMID: 40373259 DOI: 10.1200/po-24-00658] [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: 09/27/2024] [Revised: 01/16/2025] [Accepted: 03/07/2025] [Indexed: 05/17/2025] Open
Abstract
PURPOSE Patients with metastatic melanoma who progress on checkpoint inhibitors and BRAF-targeting drugs have limited therapeutic options. Up to one third of melanomas harbor at least one molecular aberration in the homologous recombination (HR) pathway, leading to HR deficiency. PATIENTS AND METHODS In this single-arm trial, we assessed the overall response rate to niraparib in patients with metastatic melanoma, harboring a genetic alteration in the HR pathway (ARID1A/B, ARID2, ATM, ATR, ATRX, BARD1, BRCA1/2, BAP1, BRIP1, CHEK2, FANCD2, MRE11A, RAD50, RAD51, RAD54B, or PALB2) who had disease progression after PD-1 blockade or BRAF/MEK inhibition if BRAF-mutant. Niraparib was administered orally at 300 mg or 200 mg daily, based on body weight and platelet count. RESULTS Fourteen patients were accrued to the trial, which was discontinued because of slow accrual. The median age was 71 years. Nine patients had an Eastern Cooperative Oncology Group performance status of 1. Eleven patients had elevated lactate dehydrogenase levels. Ten patients had nonuveal melanoma and four had uveal melanoma. Two (14%) had a partial response and seven (50%) had stable disease, with a disease control rate of 64%. The median progression-free survival was 16 weeks. Among the patients with nonuveal melanoma, two (20%) achieved partial response with a time to progression of 32 and 24 weeks, while five (50%) had stable disease lasting 16-98 weeks. None of the four patients with uveal melanoma responded. There were no unexpected adverse events related to niraparib treatment. Notably, one responder with an ARID1A mutation had detectable circulating tumor DNA at baseline, which became undetectable during treatment. CONCLUSION Despite the small sample size, our results indicate a promising signal for single agent niraparib in patients with pretreated nonuveal metastatic melanoma with HR gene mutations.
Collapse
Affiliation(s)
- Kevin B Kim
- Center for Melanoma Research and Treatment, Sutter California Pacific Medical Center, San Francisco, CA
- California Pacific Medical Center Research Institute, San Francisco, CA
| | | | - David de Semir
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Rinette W L Woo
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Anima Sharma
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Robyn Jones
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Chongshan Caressi
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Mehdi Nosrati
- Center for Melanoma Research and Treatment, Sutter California Pacific Medical Center, San Francisco, CA
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Emilia Janiczek
- Center for Melanoma Research and Treatment, Sutter California Pacific Medical Center, San Francisco, CA
| | - Julia Rivera Penafiel
- Center for Melanoma Research and Treatment, Sutter California Pacific Medical Center, San Francisco, CA
| | - Mohammed Kashani-Sabet
- Center for Melanoma Research and Treatment, Sutter California Pacific Medical Center, San Francisco, CA
- California Pacific Medical Center Research Institute, San Francisco, CA
| |
Collapse
|
41
|
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
|
42
|
Sweatman E, Bayley R, Selemane R, Higgs MR. SETD1A-dependent EME1 transcription drives PARPi sensitivity in HR deficient tumour cells. Br J Cancer 2025; 132:690-702. [PMID: 39994444 PMCID: PMC11997087 DOI: 10.1038/s41416-025-02963-0] [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: 07/10/2024] [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
|
43
|
Kwon WA, Joung JY. Precision Targeting in Metastatic Prostate Cancer: Molecular Insights to Therapeutic Frontiers. Biomolecules 2025; 15:625. [PMID: 40427518 PMCID: PMC12108645 DOI: 10.3390/biom15050625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Metastatic prostate cancer (mPCa) remains a significant cause of cancer-related mortality in men. Advances in molecular profiling have demonstrated that the androgen receptor (AR) axis, DNA damage repair pathways, and the PI3K/AKT/mTOR pathway are critical drivers of disease progression and therapeutic resistance. Despite the established benefits of hormone therapy, chemotherapy, and bone-targeting agents, mPCa commonly becomes treatment-resistant. Recent breakthroughs have highlighted the importance of identifying actionable genetic alterations, such as BRCA2 or ATM defects, that render tumors sensitive to poly-ADP ribose polymerase (PARP) inhibitors. Parallel efforts have refined imaging-particularly prostate-specific membrane antigen (PSMA) positron emission tomography-computed tomography-to detect and localize metastatic lesions with high sensitivity, thereby guiding patient selection for PSMA-targeted radioligand therapies. Multi-omics innovations, including liquid biopsy technologies, enable the real-time tracking of emergent AR splice variants or reversion mutations, supporting adaptive therapy paradigms. Nonetheless, the complexity of mPCa necessitates combination strategies, such as pairing AR inhibition with PI3K/AKT blockade or PARP inhibitors, to inhibit tumor plasticity. Immuno-oncological approaches remain challenging for unselected patients; however, subsets with mismatch repair deficiency or neuroendocrine phenotypes may benefit from immune checkpoint blockade or targeted epigenetic interventions. We present these pivotal advances, and discuss how biomarker-guided integrative treatments can improve mPCa management.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Jae Young Joung
- Department of Urology, Urological Cancer Center, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
44
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [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: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
45
|
Gui F, Jiang B, Jiang J, He Z, Tsujino T, Takai T, Arai S, Pana C, Köllermann J, Bradshaw GA, Eisert R, Kalocsay M, Fassl A, Balk SP, Kibel AS, Jia L. Acute BRCAness induction and AR pathway blockage through CDK12/7/9 degradation enhances PARP inhibitor sensitivity in prostate cancer. SCIENCE ADVANCES 2025; 11:eadu0847. [PMID: 40267193 PMCID: PMC12017310 DOI: 10.1126/sciadv.adu0847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/17/2025] [Indexed: 04/25/2025]
Abstract
Current treatments for advanced prostate cancer (PCa) primarily target the androgen receptor (AR) pathway. However, the emergence of castration-resistant prostate cancer (CRPC) and resistance to AR pathway inhibitors (APPIs) remains ongoing challenges. Here, we present BSJ-5-63, a proteolysis-targeting chimera (PROTAC) targeting cyclin-dependent kinases (CDKs) CDK12, CDK7, and CDK9, offering a multipronged approach to CRPC therapy. BSJ-5-63 degrades CDK12, diminishing BRCA1 and BRCA2 expression and inducing a sustained "BRCAness" state. This sensitizes cancer cells to PARP inhibitors (PARPis) regardless of their homologous recombination repair (HRR) status. Furthermore, CDK7 and CDK9 degradation attenuates AR signaling, enhancing its therapeutic efficacy. Preclinical studies, including both in vitro and in vivo CRPC models, demonstrate that BSJ-5-63 exerts potent antitumor activity in both AR-positive and AR-negative setting. This study introduces BSJ-5-63 as a promising therapeutic agent that addresses both DNA repair and AR signaling mechanisms, with potential benefits for a board patient population.
Collapse
Affiliation(s)
- Fu Gui
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Baishan Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jie Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Zhixiang He
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Takuya Tsujino
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Tomoaki Takai
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Seiji Arai
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Celine Pana
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Jens Köllermann
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Pathology, Frankfurt am Main, Germany
| | | | - Robyn Eisert
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Marian Kalocsay
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anne Fassl
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Steven P. Balk
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Adam S. Kibel
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Jia
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Schubart C, Tögel L, Carta MG, Hetzner P, Helbig L, Zaglas C, Ziegler M, Stöhr R, Hölsken A, Hoyer J, Ferrazzi F, Neufert C, Lettmaier S, Pavel M, Golcher H, Mueller SK, Fuchs F, Schulmeyer CE, Beckmann MW, Wullich B, Agaimy A, Reis A, Hartmann A, Meidenbauer N, Spoerl S, Haller F, Moskalev EA. Limited association between HRR gene alterations and HRD in molecular tumor board cancer samples: Who should be tested for HRD? Int J Cancer 2025. [PMID: 40278800 DOI: 10.1002/ijc.35457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/02/2024] [Accepted: 01/25/2025] [Indexed: 04/26/2025]
Abstract
Alterations in Homologous Recombination Repair (HRR) Pathway genes have been found to be associated with HR-Deficiency (HRD), which is an approved biomarker for PARP Inhibitor (PARPi) treatment. The aim of a Molecular Tumor Board (MTB) is to identify molecular alterations in cancer patients with advanced tumors that may suggest off-label treatment options. So far, few studies have analyzed the presence of HRR gene mutations and their association with HRD outside of clinical studies. Currently, no data on HRD testing in the setting of a MTB have been published. For the present study, a cohort of 237 patients encompassing 24 different tumor entities was collected from the MTB of the Comprehensive Cancer Center Erlangen-EMN. We show that an elevated Genomic Instability Score (GIS ≥42) can occur in samples with and without mutations in HRR-related genes. Overall, 38.1% of cancer samples with BRCA1/2 mutations, 10.9% of tumors with alterations in HRR genes other than BRCA1/2, and 4.3% of cancer samples without HRR gene mutations harbored an elevated GIS. Notably, our data show that various inactivating BRCA1/2 mutations are not associated with an elevated GIS. Taken together, panCancer assessment of HRD in addition to BRCA1/2 and other HRR gene mutational analysis is recommended to guide decisions regarding PARPi treatment. Further studies are needed to establish thresholds for GIS in non-ovarian cancer entities. Finally, HRD can be observed in 4.3% of BRCA1/2 and other HRR gene wildtype cancer samples, and may emerge as an independent biomarker for PARPi in the future.
Collapse
Affiliation(s)
- Christoph Schubart
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Lars Tögel
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Maria Giulia Carta
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Philip Hetzner
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lina Helbig
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Charlotte Zaglas
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maria Ziegler
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert Stöhr
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Annett Hölsken
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Juliane Hoyer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Fulvia Ferrazzi
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Clemens Neufert
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sebastian Lettmaier
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Marianne Pavel
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Henriette Golcher
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sarina K Mueller
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of ENT, Head and Neck Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Fuchs
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Carla E Schulmeyer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Department of Obstetrics and Gynecology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matthias W Beckmann
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Obstetrics and Gynecology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Bernd Wullich
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Andre Reis
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Centre for Rare Diseases Erlangen, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Norbert Meidenbauer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Internal Medicine 5, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Silvia Spoerl
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Internal Medicine 5, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Haller
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Evgeny A Moskalev
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
47
|
Jiang B, Bi Y, Chen Y, Bi J, Deng J, Zhang G. Case Report: Two cases of chemotherapy refractory aggressive variant prostate cancer with extreme durable response to PARP inhibitor. Front Oncol 2025; 15:1533627. [PMID: 40342821 PMCID: PMC12058769 DOI: 10.3389/fonc.2025.1533627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 04/02/2025] [Indexed: 05/11/2025] Open
Abstract
Background Aggressive variant prostate cancer (AVPC) represents a distinct clinical subset characterized by resistance to novel hormone therapies and an unfavorable prognosis, frequently associated with the concurrent loss of tumor suppressor genes (TSG) such as PTEN, RB1, and TP53. While the progression-free survival (PFS) and overall survival (OS) of AVPC are relatively short, the optimal first-line treatment remains unclear. Presentation In this case report, we presented two de novo AVPC cases who have ultimately benefited from the usage of PARP inhibitors. The first patient was a 64-year-old male who was diagnosed during prostate biopsy featured by mutations in PTEN, and loss of RB1, BRCA2, ATM, and FANCA. He was treated with docetaxel/albumin-bound paclitaxel and cisplatin in the first line. Second-line therapy was applied with radiotherapy and Olaparib after failure of first-line therapy, resulting in a PSA response sustained for three years. The second case was a 75-year-old male with localized neuroendocrine feature and mutations in TP53, loss of RB1 and HDAC2. He was treated with sustained ADT and chemotherapy in the first-line treatment. Radiotherapy and Fluzoparib + abiraterone was applied as subsequent treatments with a PSA response for 2 years. Conclusions These two cases demonstrating a satisfactorily durable response to PARP inhibitors indicating its clinical benefit in AVPC population with detected DNA damage response (DDR) defects. The survival improvement with PARP inhibitors observed in our clinical experiences, along with current advances in tumor sequencing provide more information on future clinical trials and explorations of innovative therapies in AVPC population.
Collapse
Affiliation(s)
- Bohao Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yifan Bi
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiming Chen
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianbin Bi
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Deng
- Third Department of Medical Oncology, The Fifth People Hospital of Shenyang, Shenyang, Liaoning, China
| | - Gejun Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
48
|
Abstract
Importance Prostate cancer is the most common nonskin cancer in men in the US, with an estimated 299 010 new cases and 35 250 deaths in 2024. Prostate cancer is the second most common cancer in men worldwide, with 1 466 680 new cases and 396 792 deaths in 2022. Observations The most common type of prostate cancer is adenocarcinoma (≥99%), and the median age at diagnosis is 67 years. More than 50% of prostate cancer risk is attributable to genetic factors; older age and Black race (annual incidence rate, 173.0 cases per 100 000 Black men vs 97.1 cases per 100 000 White men) are also strong risk factors. Recent guidelines encourage shared decision-making for prostate-specific antigen (PSA) screening. At diagnosis, approximately 75% of patients have cancer localized to the prostate, which is associated with a 5-year survival rate of nearly 100%. Based on risk stratification that incorporates life expectancy, tumor grade (Gleason score), tumor size, and PSA level, one-third of patients with localized prostate cancer are appropriate for active surveillance with serial PSA measurements, prostate biopsies, or magnetic resonance imaging, and initiation of treatment if the Gleason score or tumor stage increases. For patients with higher-risk disease, radiation therapy or radical prostatectomy are reasonable options; treatment decision-making should include consideration of adverse events and comorbidities. Despite definitive therapy, 2% to 56% of men with localized disease develop distant metastases, depending on tumor risk factors. At presentation, approximately 14% of patients have metastases to regional lymph nodes. An additional 10% of men have distant metastases that are associated with a 5-year survival rate of 37%. Treatment of metastatic prostate cancer primarily relies on androgen deprivation therapy, most commonly through medical castration with gonadotropin-releasing hormone agonists. For patients with newly diagnosed metastatic prostate cancer, the addition of androgen receptor pathway inhibitors (eg, darolutamide, abiraterone) improves survival. Use of abiraterone improved the median overall survival from 36.5 months to 53.3 months (hazard ratio, 0.66 [95% CI, 0.56-0.78]) compared with medical castration alone. Chemotherapy (docetaxel) may be considered, especially for patients with more extensive disease. Conclusions and Relevance Approximately 1.5 million new cases of prostate cancer are diagnosed annually worldwide. Approximately 75% of patients present with cancer localized to the prostate, which is associated with a 5-year survival rate of nearly 100%. Management includes active surveillance, prostatectomy, or radiation therapy, depending on risk of progression. Approximately 10% of patients present with metastatic prostate cancer, which has a 5-year survival rate of 37%. First-line therapies for metastatic prostate cancer include androgen deprivation and novel androgen receptor pathway inhibitors, and chemotherapy for appropriate patients.
Collapse
Affiliation(s)
- Ruben Raychaudhuri
- Department of Medicine, University of Washington, Seattle
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Daniel W Lin
- Department of Urology, University of Washington, Seattle
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - R Bruce Montgomery
- Department of Medicine, University of Washington, Seattle
- Fred Hutchinson Cancer Center, Seattle, Washington
- VA Puget Sound Health Care System, Seattle, Washington
| |
Collapse
|
49
|
Giri VN, Rumble RB, Yu EY, Lu K. Germline and Somatic Genomic Testing for Metastatic Prostate Cancer: ASCO Guideline Clinical Insights. JCO Oncol Pract 2025:OP2500186. [PMID: 40239123 DOI: 10.1200/op-25-00186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Veda N Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, CT
| | | | - Evan Y Yu
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA
| | - Kevin Lu
- Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung City, Taiwan
| |
Collapse
|
50
|
Zheng W, Ge Z, Wu Q, Wan H, Sun J, Nai Y, Lv C. Olaparib Combined with Anti-PD1 Enhances Immunotherapy of Gastric Cancer Via NF-κB/c-Myc/PD-L1 Signaling. Dig Dis Sci 2025:10.1007/s10620-025-09021-y. [PMID: 40237904 DOI: 10.1007/s10620-025-09021-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND PARP inhibitors, effective in BRCA-mutated cancers, show potential in gastric cancer (GC) where homologous recombination defects (e.g., BRCA1/2 mutations) are common. Olaparib, a PARP inhibitor, upregulates PD-L1, suggesting synergy with PD-1 inhibitors for enhanced GC therapy. METHODS Using CCK-8 screening of 867 drugs, olaparib demonstrated potent GC cell inhibition. Western blot and qRT-PCR assessed PD-L1, c-MYC, COX-2, and NF-κB pathway proteins (p65/p-p65). Functional assays (Transwell, wound healing, colony formation) evaluated olaparib's effects on GC cell proliferation, migration, and invasion. A GC mouse model tested olaparib combined with anti-PD1. TCGA and Kaplan-Meier analyzed PARP expression-prognosis correlations. RESULTS Olaparib suppressed GC cell proliferation, migration, and invasion in vitro. Western blot revealed upregulated c-MYC, COX-2, p65, p-p65, and PD-L1, confirmed by qRT-PCR for PD-L1. Low PARP expression correlated with better GC patient survival. In vivo, olaparib synergized with anti-PD1 to enhance tumor suppression. CONCLUSION Olaparib activates the NF-κB/c-MYC pathway to elevate PD-L1, supporting its combination with PD-1 inhibitors as a promising GC therapeutic strategy.
Collapse
Affiliation(s)
- Wubin Zheng
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Zhifa Ge
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Qingwei Wu
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Haoyue Wan
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Junjie Sun
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| | - Yongjun Nai
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China.
| | - Chengyu Lv
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, 68 Changle Rd., Nanjing, 210006, Jiangsu, China
| |
Collapse
|