Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (136)

Search Parameters:
Keywords = hormone-sensitive prostate cancer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 357 KiB  
Article
Apalutamide Monotherapy in Metastatic Hormone-Sensitive Prostate Cancer: A Viable Alternative to First-Generation Anti-Androgen Agents to Avoid the Flare Phenomenon and an Effective Treatment for Achieving Early PSA Response
by Gaetano Facchini, Andrea D’Arienzo, Antonella Nicastro, Fabiano Flauto, Michela Izzo, Liliana Montella, Filippo Riccardo, Giovanni Maria Fusco, Francesco Trama, Giovanni Di Lauro, Giuseppe Di Costanzo, Anna Giacoma Tucci, Francesca Iasiello, Lorena Di Lorenzo, Salvatore Maddaluno, Carmela Liguori, Rita Assante di Cupillo, Paola Coppola, Angela Minissale, Maria Teresa Di Nardo, Luigi Formisano, Erika Martinelli, Giuliana Ciappina, Salvatore Pisconti, Massimiliano Berretta and Chiara Barracoadd Show full author list remove Hide full author list
Cancers 2025, 17(15), 2573; https://doi.org/10.3390/cancers17152573 - 5 Aug 2025
Abstract
Background/Objectives: Androgen deprivation therapy (ADT) is the mainstay of prostate cancer treatment, especially in advanced disease. In particular, the gonadotropin-releasing hormone agonists (aGnRH) reduce the production of gonadotropin and, therefore, of testosterone. In about 10% of patients, the non-pulsatile stimulation of GnRH receptor [...] Read more.
Background/Objectives: Androgen deprivation therapy (ADT) is the mainstay of prostate cancer treatment, especially in advanced disease. In particular, the gonadotropin-releasing hormone agonists (aGnRH) reduce the production of gonadotropin and, therefore, of testosterone. In about 10% of patients, the non-pulsatile stimulation of GnRH receptor initially causes a surge in LH and testosterone, defined as the “flare-up phenomenon”, leading to increased bone pain, spinal cord compression, bladder outlet obstruction and cardiovascular issues. To mitigate this effect, combining a first-generation antiandrogen agent (FGA) with aGnRH is recommended. However, second-generation anti-androgens, such as apalutamide, bind selectively and irreversibly to the androgen receptor (AR), exhibiting a more efficient inhibition of the AR pathway. Methods: This is a descriptive retrospective study of 27 patients (pts) with mHSPC, treated at a single center (“Santa Maria delle Grazie” Hospital in Pozzuoli, ASL Napoli 2 Nord, Italy) between June 2022 and April 2024. Patients received apalutamide monotherapy for 14 days followed by continuous combination with aGnRH plus apalutamide. Serum PSA and testosterone levels were measured at baseline, at day 14 (after 13 days of apalutamide monotherapy), at day 28 (after an additional 15 days of apalutamide plus a aGnRH), and at day 60. Results: PSA levels decreased from a mean of 45.2 (±63.1) ng/mL at baseline to a mean of 12.6 (±23.4) ng/mL at day 14 and to 3.3 ng/mL (±6.0) at day 28 of treatment. After 14 days of apalutamide monotherapy, 21 patients (77.8%) achieved a >50% PSA reduction and 4 (14.8%) a >90% PSA reduction. The number of patients with undetectable PSA was one (3.7%) at day 14, two (7.4%) at day 28, and nine (33.3%) at day 60. The mean serum testosterone levels were 6.56 (±4.46) ng/mL at baseline, 6.58 (±4.42) ng/mL at day 14, and 2.40 (± 3.38) ng/mL at day 28. No significant difference in PSA and testosterone level reduction during treatment emerged between subgroups of patients with low- vs. high-volume disease. Conclusions: Apalutamide alone is a viable option for mitigating the flare-up phenomenon, avoiding first generation anti-androgen therapy, and it can achieve rapid and deep biochemical control. Full article
(This article belongs to the Special Issue Advances in Therapeutic Strategies for Prostate Cancer)
Show Figures

Figure 1

15 pages, 534 KiB  
Review
Evolving Treatment Paradigms in Metastatic Hormone-Sensitive Prostate Cancer: Expert Narrative Review
by Vineet Talwar, Kaushal Kalra, Akhil Kapoor, P. S. Dattatreya, Amit Joshi, Krishna Chaitanya, M. V. Chandrakanth, Atul Batra, Krishna Prasad, Nikhil Haridas and Nilesh Lokeshwar
Curr. Oncol. 2025, 32(8), 437; https://doi.org/10.3390/curroncol32080437 - 5 Aug 2025
Abstract
The treatment landscape of metastatic hormone-sensitive prostate cancer (mHSPC) has transformed significantly with the advent of triplet therapy involving androgen deprivation therapy (ADT), docetaxel, and androgen receptor signalling inhibitors (ARSIs). While clinical guidelines increasingly support early intensification, real-world practice remains challenged by patient [...] Read more.
The treatment landscape of metastatic hormone-sensitive prostate cancer (mHSPC) has transformed significantly with the advent of triplet therapy involving androgen deprivation therapy (ADT), docetaxel, and androgen receptor signalling inhibitors (ARSIs). While clinical guidelines increasingly support early intensification, real-world practice remains challenged by patient heterogeneity, evolving evidence, and limited consensus on treatment sequencing. This narrative review integrates evidence from landmark trials, clinical guidelines, and expert insights from oncologists managing mHSPC in India. Findings affirm that triplet therapy, particularly with darolutamide, improves survival in high-volume disease and underscores the need for personalized treatment based on disease burden, comorbidities, and genomic profiles. The review also highlights gaps in real-world data, sequencing strategies, and biomarker-driven therapy, reinforcing the need for precision medicine and locally relevant evidence to guide treatment. Ultimately, optimizing mHSPC management requires harmonizing guideline-based approaches with individualized, real-world decision making to improve patient outcomes. Full article
(This article belongs to the Section Genitourinary Oncology)
Show Figures

Figure 1

12 pages, 920 KiB  
Article
Apalutamide and Stereotactic Body Radiotherapy in Metastatic Hormone-Sensitive Prostate Cancer: Multicenter Real-World Study
by Juan A. Encarnación, Virginia Morillo Macías, Isabel De la Fuente Muñoz, Violeta Derrac Soria, Luis Fernández Fornos, María Albert Antequera, Osamah Amr Rey, Vicente García Martínez, José L. Alonso-Romero and Raquel García Gómez
Cancers 2025, 17(13), 2216; https://doi.org/10.3390/cancers17132216 - 2 Jul 2025
Viewed by 577
Abstract
Background: The management of metastatic hormone-sensitive prostate cancer (mHSPC) has evolved with the integration of androgen receptor signaling inhibitors (ARSIs) and metastasis-directed therapies (MDTs). Stereotactic body radiotherapy (SBRT) offers precise local control, yet real-world data on its combination with apalutamide remain limited. Methods: [...] Read more.
Background: The management of metastatic hormone-sensitive prostate cancer (mHSPC) has evolved with the integration of androgen receptor signaling inhibitors (ARSIs) and metastasis-directed therapies (MDTs). Stereotactic body radiotherapy (SBRT) offers precise local control, yet real-world data on its combination with apalutamide remain limited. Methods: We conducted a multicenter retrospective cohort study including 134 patients with mHSPC treated with apalutamide and SBRT between February 2021 and December 2024. The primary endpoints were progression-free survival (PFS), local control (LC), and treatment safety. PSA kinetics and radiologic response were evaluated, and outcomes were analyzed according to PSA thresholds and treatment timing. Results: Most patients (93.3%) had low-volume disease; 97.1% presented with ≤5 metastases. At a median follow-up of 28 months, LC was 99.3% and 95.5% of patients were progression-free. Complete radiological response was achieved in 87.5% of patients, and 68.4% attained ultralow PSA levels (≤0.02 ng/mL). Undetectable PSA and radiologic complete response were independently associated with improved PFS (p = 0.010 and p = 0.011, respectively). Treatment was well tolerated, with grade ≥3 toxicity occurring in only 2.2% of patients. Conclusions: The combination of apalutamide and SBRT in mHSPC is associated with high local and systemic disease control and minimal toxicity in a real-world setting. This approach may delay systemic treatment intensification and the onset of castration resistance. Prospective studies are warranted to confirm these findings. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Cancer Development and Metastasis)
Show Figures

Figure 1

17 pages, 4423 KiB  
Article
Multivariate Framework of Metabolism in Advanced Prostate Cancer Using Whole Abdominal and Pelvic Hyperpolarized 13C MRI—A Correlative Study with Clinical Outcomes
by Hsin-Yu Chen, Ivan de Kouchkovsky, Robert A. Bok, Michael A. Ohliger, Zhen J. Wang, Daniel Gebrezgiabhier, Tanner Nickles, Lucas Carvajal, Jeremy W. Gordon, Peder E. Z. Larson, John Kurhanewicz, Rahul Aggarwal and Daniel B. Vigneron
Cancers 2025, 17(13), 2211; https://doi.org/10.3390/cancers17132211 - 1 Jul 2025
Cited by 1 | Viewed by 534 | Correction
Abstract
Background: Most of the existing hyperpolarized (HP) 13C MRI analyses use univariate rate maps of pyruvate-to-lactate conversion (kPL), and radiomic-style multiparametric models extracting complex, higher-order features remain unexplored. Purpose: To establish a multivariate framework based on whole abdomen/pelvis HP 13 [...] Read more.
Background: Most of the existing hyperpolarized (HP) 13C MRI analyses use univariate rate maps of pyruvate-to-lactate conversion (kPL), and radiomic-style multiparametric models extracting complex, higher-order features remain unexplored. Purpose: To establish a multivariate framework based on whole abdomen/pelvis HP 13C-pyruvate MRI and evaluate the association between multiparametric features of metabolism (MFM) and clinical outcome measures in advanced and metastatic prostate cancer. Methods: Retrospective statistical analysis was performed on 16 participants with metastatic or local-regionally advanced prostate cancer prospectively enrolled in a tertiary center who underwent HP-pyruvate MRI of abdomen or pelvis between November 2020 and May 2023. Five patients were hormone-sensitive and eleven were castration-resistant. GMP-grade [1-13C]pyruvate was polarized using a 5T clinical-research DNP polarizer, and HP MRI used a set of flexible vest-transmit, array-receive coils, and echo-planar imaging sequences. Three basic metabolic maps (kPL, pyruvate summed-over-time, and mean pyruvate time) were created by semi-automatic segmentation, from which 316 MFMs were extracted using an open-source, radiomic-compliant software package. Univariate risk classifier was constructed using a biologically meaningful feature (kPL,median), and the multivariate classifier used a two-step feature selection process (ranking and clustering). Both were correlated with progression-free survival (PFS) and overall survival (OS) (median follow-up = 22.0 months) using Cox proportional hazards model. Results: In the univariate analysis, patients harboring tumors with lower-kPL,median had longer PFS (11.2 vs. 0.5 months, p < 0.01) and OS (NR vs. 18.4 months, p < 0.05) than their higher-kPL,median counterparts. Using a hypothesis-generating, age-adjusted multivariate risk classifier, the lower-risk subgroup also had longer PFS (NR vs. 2.4 months, p < 0.002) and OS (NR vs. 18.4 months, p < 0.05). By contrast, established laboratory markers, including PSA, lactate dehydrogenase, and alkaline phosphatase, were not significantly associated with PFS or OS (p > 0.05). Key limitations of this study include small sample size, retrospective study design, and referral bias. Conclusions: Risk classifiers derived from select multiparametric HP features were significantly associated with clinically meaningful outcome measures in this small, heterogeneous patient cohort, strongly supporting further investigation into their prognostic values. Full article
Show Figures

Figure 1

20 pages, 3183 KiB  
Article
Clinical and Transcriptomic Characterization of Metastatic Hormone-Sensitive Prostate Cancer Patients with Low PTEN Expression
by Marta Garcia de Herreros, Natalia Jiménez, Joan Padrosa, Caterina Aversa, Laura Ferrer-Mileo, Samuel García-Esteve, Leonardo Rodríguez-Carunchio, Isabel Trias, Laia Fernández-Mañas, Mercedes Marín-Aguilera, Mariana Altamirano, Manuel Mazariegos, Albert Font, Alejo Rodriguez-Vida, Miguel Ángel Climent, Sara Cros, Isabel Chirivella, Mariona Figols, Núria Sala-González, Vicenç Ruiz de Porras, Juan Carlos Pardo, Aleix Prat, Òscar Reig and Begoña Melladoadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2025, 26(13), 6244; https://doi.org/10.3390/ijms26136244 - 28 Jun 2025
Viewed by 810
Abstract
Alterations in the PTEN tumor suppressor gene are common in prostate cancer. They have been associated with a more aggressive disease and poor outcomes and potential benefit of targeted therapies. The purpose of this work is to study the clinical and transcriptional landscapes [...] Read more.
Alterations in the PTEN tumor suppressor gene are common in prostate cancer. They have been associated with a more aggressive disease and poor outcomes and potential benefit of targeted therapies. The purpose of this work is to study the clinical and transcriptional landscapes associated to low PTEN mRNA expression in metastatic hormone-sensitive prostate cancer (mHSPC) patients. A multicenter biomarker ambispective study was performed in mHSPC patients. PTEN mRNA expression was assessed by nCounter in formalin-fixed paraffin-embedded tumor samples. PTENlow status was defined by a previously validated cut-off and was correlated with castration-resistant prostate cancer (CRPC)-free survival (CRPC-FS) (primary endpoint) and overall survival (OS). RNA-Seq was performed to molecularly characterize PTENlow vs. PTENwt tumors. A total of 380 patients were included, 350 eligible. PTENlow was observed in 28.2% of patients and was independently associated with shorter CRPC-FS (HR 1.6, 95% CI 1.2–2.1, p = 0.002) and OS (HR 1.5, 95% CI 1.1–2, p = 0.014). PTENlow tumors showed overexpression of neuroendocrine, cell cycle, and DNA repair gene signatures, reduced expression of the androgen receptor pathway, and a distinct immune microenvironment. Using microarray data from the CHAARTED trial, we developed a PTEN-low related signature, independently associated with CRPC-FS (HR 1.5, 95% CI 1–2.3, p = 0.036) and OS (HR 1.9, C1 1.2–2.9, p = 0.005), and identified targets for potential therapies in PTEN-altered tumors. We conclude that PTENlow correlates with an aggressive clinical outcome in mHSPC patients and is associated with a unique transcriptional profile. These findings further support the investigation of novel therapeutic strategies for patients with PTEN alterations. Full article
(This article belongs to the Special Issue Molecular Research on Prostate Cancer)
Show Figures

Graphical abstract

16 pages, 735 KiB  
Article
Integrating Aggressive-Variant Prostate Cancer-Associated Tumor Suppressor Gene Status with Clinical Variables to Refine Prognosis and Predict Androgen Receptor Pathway Inhibitor Response in Metastatic Hormone-Sensitive Setting
by Martino Pedrani, Giuseppe Salfi, Sara Merler, Irene Testi, Chiara Maria Agrippina Clerici, Giovanna Pecoraro, Luis Castelo-Branco, Fabio Turco, Luigi Tortola, Ursula Vogl, Silke Gillessen, Jean-Philippe Theurillat, Thomas Zilli and Ricardo Pereira Mestre
Int. J. Mol. Sci. 2025, 26(11), 5309; https://doi.org/10.3390/ijms26115309 - 31 May 2025
Viewed by 727
Abstract
Alterations in aggressive-variant prostate cancer-associated tumor suppressor genes (AVPC-TSG: TP53, RB1, PTEN) are related with androgen insensitivity and aggressive disease. However, their prognostic and predictive role in metastatic hormone-sensitive prostate cancer (mHSPC) is unclear. This single-center retrospective study assesses the [...] Read more.
Alterations in aggressive-variant prostate cancer-associated tumor suppressor genes (AVPC-TSG: TP53, RB1, PTEN) are related with androgen insensitivity and aggressive disease. However, their prognostic and predictive role in metastatic hormone-sensitive prostate cancer (mHSPC) is unclear. This single-center retrospective study assesses the value of AVPC-TSG alterations in refining prognosis and predicting the response to androgen receptor pathway inhibitors (ARPIs) in mHSPC. We included 158 patients with genomic tumor sequencing undergoing treatment for mHSPC between 2013 and 2023. We compared patients with AVPC-TSGalt tumors (≥1 alteration in TP53, RB1, or PTEN/PI3K/AKT pathway genes) to those with AVPC-TSGwt tumors (i.e., without alterations in AVPC-TSG). Cox analyses were performed for progression-free survival (PFS) and overall survival (OS). AVPC-TSGwt status was associated with improved PFS and OS in both univariate and multivariate (MV) analyses (MV PFS: HR 0.58, 95%CI: 0.38–0.89, p = 0.012; MV OS: HR 0.48, 95%CI: 0.26–0.91, p = 0.025). AVPC-TSGalt mHSPC patients seemed to derive no PFS benefit from ARPI addition (PFS: HR 1.13, 95%CI: 0.58–2.19, p = 0.721), while AVPC-TSGwt mHSPC patients did (PFS: HR 0.51, 95%CI: 0.28–0.93 p = 0.029). Integrating AVPC-TSG status with CHAARTED volume criteria, we identified three distinct subgroups: “good risk” (AVPC-TSGwt low volume), “intermediate risk” (either AVPC-TSGalt low volume or AVPC-TSGwt high volume), and “poor risk” (AVPC-TSGalt high volume) with median PFS of 46.8, 28.2, and 15.7 months, respectively. Only the “intermediate risk” subgroup seemed to derive PFS benefit from ARPI addition (HR 0.36, 95%CI: 0.19–0.70, p = 0.002). AVPC-TSG status assessment refines prognosis and may predict PFS benefits of ARPIs in mHSPC. AVPC-TSGalt mHSPC patients should be considered for clinical trials as they may not benefit from current standard approaches. Full article
(This article belongs to the Special Issue Molecular Research in Prostate Cancer)
Show Figures

Figure 1

17 pages, 2678 KiB  
Article
A Multiparametric MRI and Baseline-Clinical-Feature-Based Dense Multimodal Fusion Artificial Intelligence (MFAI) Model to Predict Castration-Resistant Prostate Cancer Progression
by Dianning He, Haoming Zhuang, Ying Ma, Bixuan Xia, Aritrick Chatterjee, Xiaobing Fan, Shouliang Qi, Wei Qian, Zhe Zhang and Jing Liu
Cancers 2025, 17(9), 1556; https://doi.org/10.3390/cancers17091556 - 3 May 2025
Viewed by 679
Abstract
Objectives: The primary objective of this study was to identify whether patients with prostate cancer (PCa) could progress to denervation-resistant prostate cancer (CRPC) after 12 months of hormone therapy. Methods: A total of 96 PCa patients with baseline clinical data who underwent multiparametric [...] Read more.
Objectives: The primary objective of this study was to identify whether patients with prostate cancer (PCa) could progress to denervation-resistant prostate cancer (CRPC) after 12 months of hormone therapy. Methods: A total of 96 PCa patients with baseline clinical data who underwent multiparametric magnetic resonance imaging (MRI) between September 2018 and September 2022 were included in this retrospective study. Patients were classified as progressing or not progressing to CRPC on the basis of their outcome after 12 months of hormone therapy. A dense multimodal fusion artificial intelligence (Dense-MFAI) model was constructed by incorporating a squeeze-and-excitation block and a spatial pyramid pooling layer into a dense convolutional network (DenseNet), as well as integrating the eXtreme Gradient Boosting machine learning algorithm. The accuracy, sensitivity, specificity, positive predictive value, negative predictive value, receiver operating characteristic curves, area under the curve (AUC) and confusion matrices were used as classification performance metrics. Results: The Dense-MFAI model demonstrated an accuracy of 94.2%, with an AUC of 0.945, when predicting the progression of patients with PCa to CRPC after 12 months of hormone therapy. The experimental validation demonstrated that combining radiomics feature mapping with baseline clinical characteristics significantly improved the model’s classification performance, confirming the importance of multimodal data. Conclusions: The Dense-MFAI model proposed in this study has the ability to more accurately predict whether a PCa patient could progress to CRPC. This model can assist urologists in developing the most appropriate treatment plan and prognostic measures. Full article
(This article belongs to the Special Issue MRI in Prostate Cancer)
Show Figures

Figure 1

42 pages, 3927 KiB  
Review
Precision Targeting in Metastatic Prostate Cancer: Molecular Insights to Therapeutic Frontiers
by Whi-An Kwon and Jae Young Joung
Biomolecules 2025, 15(5), 625; https://doi.org/10.3390/biom15050625 - 27 Apr 2025
Cited by 1 | Viewed by 1657
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue Prostate Cancer Biomarkers and Therapeutics)
Show Figures

Figure 1

16 pages, 624 KiB  
Review
Advances in Current Treatment Paradigms for Metastatic Hormone-Sensitive Prostate Cancer
by Shayan Smani, Julien DuBois, Ismail Ajjawi, Nishan Sohoni, Ankur U. Choksi, Soum D. Lokeshwar, Isaac Y. Kim and Joseph F. Renzulli
J. Clin. Med. 2025, 14(8), 2565; https://doi.org/10.3390/jcm14082565 - 8 Apr 2025
Viewed by 2276
Abstract
Metastatic hormone-sensitive prostate cancer (mHSPCa) presents de novo or represents significant disease progression and requires systemic treatment. However, progression to castration resistance is inevitable. The treatment landscape has evolved with the introduction of intensified systemic therapy, including androgen deprivation therapy (ADT) combined with [...] Read more.
Metastatic hormone-sensitive prostate cancer (mHSPCa) presents de novo or represents significant disease progression and requires systemic treatment. However, progression to castration resistance is inevitable. The treatment landscape has evolved with the introduction of intensified systemic therapy, including androgen deprivation therapy (ADT) combined with either androgen receptor signaling inhibitors (ARSIs) or cytotoxic chemotherapy (doublet therapy) or combined therapy with both agents (triplet therapy). Landmark trials such as CHAARTED, STAMPEDE, LATITUDE, ENZAMET, and TITAN have established combination therapies as the standard of care, demonstrating significant overall survival benefits. More recently, triplet therapy—integrating ADT, docetaxel, and an ARSI—has emerged as an effective approach, particularly in high-volume metastatic disease, as supported by ARASENS and PEACE-1. Advances in imaging, such as PSMA PET-CT, have improved disease detection, allowing earlier detection of metastasis and appropriate therapy. Similarly, genomic profiling has enabled biomarker-driven, personalized treatment strategies. The role of treatment of the primary tumor, by either radiation therapy or cytoreductive prostatectomy, in low-volume disease continues to be explored. As novel therapies, targeted agents, and immunotherapies undergo investigation, optimizing treatment selection based on disease burden, molecular characteristics, and patient factors will be essential. The future of mHSPCa management lies in multidisciplinary, precision-based approaches to improve patient outcomes while balancing treatment efficacy and tolerability. Full article
(This article belongs to the Special Issue Evolving Therapies in Metastatic Prostate Cancer)
Show Figures

Figure 1

16 pages, 1290 KiB  
Systematic Review
Metastasis-Directed Therapy in Oligometastatic Prostate Cancer: Biological Rationale and Systematic Review of Published Data
by Francesco Fiorica, Teodoro Sava, Jacopo Giuliani, Umberto Tebano, Giuseppe Napoli, Antonella Franceschetto, Emilia Durante, Ilaria Campisi, Erica Palesandro, Fabio Turco, Consuelo Buttigliero, Fernando Munoz and Marcello Tucci
Cancers 2025, 17(8), 1256; https://doi.org/10.3390/cancers17081256 - 8 Apr 2025
Viewed by 1017
Abstract
Introduction: Metastasis-directed therapy (MDT) alone may be effective in preventing disease progression and positively affecting overall survival (OS) in oligometastatic prostate cancer (OMPC). Objective: We systematically reviewed the current literature to analyse the biological rationale for integrating MDT into treatment strategies for OMPC [...] Read more.
Introduction: Metastasis-directed therapy (MDT) alone may be effective in preventing disease progression and positively affecting overall survival (OS) in oligometastatic prostate cancer (OMPC). Objective: We systematically reviewed the current literature to analyse the biological rationale for integrating MDT into treatment strategies for OMPC and investigate the current evidence on its role in OMPC. Evidence acquisition: MEDLINE/PUBMED and the EMBASE Database were systematically searched to identify eligible reports published up to January 2024. The proceedings of the European Society for Radiotherapy and Oncology, European Society of Medical Oncology, American Society for Radiation Oncology, American Society of Clinical Oncology, European Uro-Oncology Group, and American Urological Association annual meetings were analysed. Results: Eighteen studies published between 2014 and 2024 were selected for the analysis. The studies included 1058 patients treated with metastasis-directed radiotherapy. No statistically significant differences were found in terms of treatment-escalation-free survival between hormone-naïve patients treated with MDT alone and those treated with MDT and hormonal manipulation. By contrast, the combination treatment significantly increased both 2 year and 4 year disease-progression-free survival (DPFS) rates (p-values < 0.00001 and 0.006, respectively). In patients with castration-sensitive disease treated with MDT alone, the estimated 2 year and 4 year OS rates were 96.4% (95% confidence interval [CI], 92.9–100%) and 89.1% (95% CI, 82.3–96.5%), respectively. The estimated 2 year and 4 year overall survival rates in the combination treatment group were 86.1% (95% CI 79.2–93.7%) and 74.8% (95% CI 64.6.3–86.5%), respectively. Conclusions: MDT alone is associated with promising outcomes in OMPC and represents a valuable, valid, and often preferable strategy. Combined with ADT improves significantly disease-progression-free survival, but its impact on overall survival remains uncertain. Given these findings, the decision to incorporate ADT should be tailored to individual patient characteristics and clinical context. Future research should integrate biomarker-based approaches to optimise MDT use and select the best candidates for a multimodal approach. Full article
(This article belongs to the Section Cancer Metastasis)
Show Figures

Figure 1

18 pages, 3488 KiB  
Article
Combined Therapy Targeting MET and Pro-HGF Activation Shows Significant Therapeutic Effect Against Liver Metastasis of CRPC
by Shoichi Kimura, Satoshi Iwano, Takahiro Akioka, Takahiro Kuchimaru, Makiko Kawaguchi, Tsuyoshi Fukushima, Yuichiro Sato, Hiroaki Kataoka, Toshiyuki Kamoto, Shoichiro Mukai and Atsuro Sawada
Int. J. Mol. Sci. 2025, 26(5), 2308; https://doi.org/10.3390/ijms26052308 - 5 Mar 2025
Viewed by 1014
Abstract
The liver is the most lethal metastatic site in castration-resistant prostate cancer (CRPC). Overexpression of MET protein has been reported in CRPC, and MET is an important driver gene in androgen-independent CRPC cells. Mouse CRPC cell line CRTC2 was established by subcutaneous injection [...] Read more.
The liver is the most lethal metastatic site in castration-resistant prostate cancer (CRPC). Overexpression of MET protein has been reported in CRPC, and MET is an important driver gene in androgen-independent CRPC cells. Mouse CRPC cell line CRTC2 was established by subcutaneous injection of hormone-sensitive PC cells (TRAMP-C2) in castrated nude mice. CRCT2/luc2 cells were injected into the spleen of castrated nude mice, and liver metastasis was confirmed at 2 weeks post-injection. We administered MET inhibitor (MET-I) and HGF activator inhibitor (HGFA-I) to this liver metastasis model and assessed the therapeutic effect. After intrasplenic injection, CRTC2 showed a higher incidence of liver metastasis whereas no metastasis was observed in TRAMP-C2. Microarray analysis revealed increased expression of HGF, MET, and HPN, HGFAC (encoding HGF activating proteases) in liver metastasis. Proliferation of CRCT2 was significantly inhibited by co-administration of MET-I and HGFA-I by in vitro analysis with HGF-enriched condition. In an analysis of the mouse model, the combination-therapy group showed the strongest reduction for liver metastasis. Immunohistochemical staining also revealed the strongest decrease in phosphorylation of MET in the combination-therapy group. Co-culture with HGF-expressed mouse fibroblasts showed attenuation of the inhibitory effect of MET-I; however, additional HGFA-I overcame the resistance. We established an androgen-independent CRPC cell line, CRTC2, and liver metastasis model in mice. Significant effect was confirmed by combined treatment of MET-I and HGFA-I by in vitro and in vivo analysis. The results suggested the importance of combined treatment with both MET- and HGF-targeting agents in the treatment of HGF-enriched conditions including liver metastasis. Full article
(This article belongs to the Special Issue Molecular Research of Therapeutic Target Enzymes)
Show Figures

Figure 1

12 pages, 661 KiB  
Article
Multicenter Real-World Study: 432 Patients with Apalutamide in Metastatic Hormone-Sensitive Prostate Cancer
by Juan A. Encarnación Navarro, Virginia Morillo Macías, María Borrás Calbo, Isabel De la Fuente Muñoz, Antonio Lozano Martínez, Vicente García Martínez, Luis Fernández Fornos, Miriam Guijarro Roche, Osamah Amr Rey and Raquel García Gómez
Curr. Oncol. 2025, 32(3), 119; https://doi.org/10.3390/curroncol32030119 - 21 Feb 2025
Cited by 1 | Viewed by 998
Abstract
Background: The management of metastatic hormone-sensitive prostate cancer (mHSPC) has evolved significantly in recent years due to the introduction of androgen receptor-targeted agents (ARTAs). When used alongside androgen deprivation therapy (ADT), these treatments have shown improved oncological results and enhanced survival rates for [...] Read more.
Background: The management of metastatic hormone-sensitive prostate cancer (mHSPC) has evolved significantly in recent years due to the introduction of androgen receptor-targeted agents (ARTAs). When used alongside androgen deprivation therapy (ADT), these treatments have shown improved oncological results and enhanced survival rates for patients with this condition. Objectives: The objective of this study was to describe the decline in prostate-specific antigen (PSA), the oncological outcomes, and the toxicity profile of mHSPC patients treated with apalutamide. Materials and Methods: Clinical data obtained from seven national hospitals were utilized between March 2021 and July 2024. PSA responses were collected at 3, 6, 12, 18, and 24 months, along with adverse effects reported by patients, dose reductions, or drug discontinuations. The association between PSA decline and progression-free survival (PFS) was evaluated with respect to metastasis volume, location, and timing of diagnosis. Results: A total of 432 patients were included, of whom 40% were de novo cases, and the greater part were classified as M1b. After one year, a reduction of more than 90% in PSA levels was observed in 88.2% of cases, with undetectable levels (≤0.2 ng/mL) achieved in 81.7% of them. The drug was discontinued in 76 patients (15.6%), with adverse effects reported in 7.8% (grade 3) and 1.9% (grade 4). Regarding PSA levels <0.02 ng/mL, promising results were observed, with ultralow PSA (UL2) achieved in 43% of cases at 6 months. Conclusions: This study revealed strong oncological outcomes, with rapid and profound PSA declines and drug safety consistent with emerging evidence. The distinctive finding of this study underscores the importance of a rapid and profound response (UL PSA) as a predictor of better oncological outcomes. Full article
(This article belongs to the Section Genitourinary Oncology)
Show Figures

Figure 1

15 pages, 2086 KiB  
Case Report
Salvage Ultrasound-Guided Robot-Assisted Video-Endoscopic Inguinal Lymphadenectomy (RAVEIL) as a Metastasis-Directed Therapy (MDT) in Oligoprogressive Metastatic Castration-Resistant Prostate Cancer (mCRPC): A Case Report and Review of the Literature
by Rafał B. Drobot, Marcin Lipa and Artur A. Antoniewicz
Curr. Oncol. 2025, 32(2), 115; https://doi.org/10.3390/curroncol32020115 - 18 Feb 2025
Viewed by 1361
Abstract
Background: Metastatic castration-resistant prostate cancer (mCRPC) remains challenging due to progression despite androgen deprivation therapy (ADT). Current treatments, including androgen receptor-targeted agents, chemotherapy, bone-targeted agents, and PARP inhibitors, extend survival but face challenges, such as resistance, adverse effects, and limited durability. Metastasis-directed [...] Read more.
Background: Metastatic castration-resistant prostate cancer (mCRPC) remains challenging due to progression despite androgen deprivation therapy (ADT). Current treatments, including androgen receptor-targeted agents, chemotherapy, bone-targeted agents, and PARP inhibitors, extend survival but face challenges, such as resistance, adverse effects, and limited durability. Metastasis-directed therapies (MDTs), such as stereotactic ablative radiotherapy (SABR), show promise in oligometastatic disease, but their role in oligoprogressive mCRPC is unclear. Salvage lymphadenectomy is rarely pursued due to invasiveness and limited data. This is the first report of robotic surgery as an MDT in this setting, demonstrating the potential of salvage robot-assisted video-endoscopic inguinal lymphadenectomy (RAVEIL) to manage oligoprogressive mCRPC and delay systemic progression. Methods: A 47-year-old male with metastatic hormone-sensitive prostate cancer (Gleason 10) underwent ADT, docetaxel chemotherapy, and radical retropubic prostatectomy with super-extended pelvic and retroperitoneal lymphadenectomy. Upon progression to oligoprogressive mCRPC, 68Ga-PSMA PET/CT detected a single metastatic inguinal lymph node. Salvage RAVEIL was performed using the da Vinci X™ Surgical System, guided by preoperative ultrasound mapping. Results: Histopathology confirmed metastasis in one of the eight excised lymph nodes. The patient achieved undetectable PSA levels and prolonged biochemical progression-free survival. Minor complications (lymphorrhea, cellulitis) resolved without sequelae. No further progression was observed for over 14 months. Conclusions: This case highlights RAVEIL as a viable MDT option for oligoprogressive mCRPC, potentially extending progression-free intervals while minimizing systemic treatment. Full article
(This article belongs to the Section Genitourinary Oncology)
Show Figures

Figure 1

15 pages, 1360 KiB  
Review
TP53 Deficiency in the Natural History of Prostate Cancer
by Heidemarie Ofner, Gero Kramer, Shahrokh F. Shariat and Melanie R. Hassler
Cancers 2025, 17(4), 645; https://doi.org/10.3390/cancers17040645 - 14 Feb 2025
Cited by 1 | Viewed by 2186
Abstract
Prostate cancer remains a leading cause of cancer-related mortality in men, with advanced stages posing significant treatment challenges due to high morbidity and mortality. Among genetic alterations, TP53 mutations are among the most prevalent in cancers and are strongly associated with poor clinical [...] Read more.
Prostate cancer remains a leading cause of cancer-related mortality in men, with advanced stages posing significant treatment challenges due to high morbidity and mortality. Among genetic alterations, TP53 mutations are among the most prevalent in cancers and are strongly associated with poor clinical outcomes and therapeutic resistance. This review investigates the role of TP53 mutations in prostate cancer progression, prognosis, and therapeutic development. A comprehensive analysis of preclinical and clinical studies was conducted to elucidate the molecular mechanisms, clinical implications, and potential therapeutic approaches associated with TP53 alterations in prostate cancer. TP53 mutations are highly prevalent in advanced stages, contributing to genomic instability, aggressive tumor phenotypes, and resistance to standard treatments. Emerging evidence supports the utility of liquid biopsy techniques, such as circulating tumor DNA analysis, for detecting TP53 mutations, providing prognostic value and facilitating early intervention strategies. Novel therapeutic approaches targeting TP53 have shown promise in preclinical settings, but their clinical efficacy requires further validation. Overall, TP53 mutations represent a critical biomarker for disease progression and therapeutic response in prostate cancer. Advances in detection methods and targeted therapies hold significant potential to improve outcomes for patients with TP53-mutated prostate cancer. Further research is essential to integrate TP53-based strategies into routine clinical practice. Full article
(This article belongs to the Special Issue The Role of Genes in Prostate Cancer)
Show Figures

Figure 1

12 pages, 1937 KiB  
Article
Cell Models of Castration Resistant and High Dose Testosterone-Resistant Prostate Cancer Recapitulate the Heterogeneity of Response Observed in Clinical Practice
by Laura S. Graham, Lih-Jen Su, Andrew Nicklawsky, Frances Xiuyan Feng, David Orlicky, Joseph Petraccione, Maren Salzmann-Sullivan, Steven K. Nordeen and Thomas W. Flaig
Cancers 2025, 17(4), 593; https://doi.org/10.3390/cancers17040593 - 10 Feb 2025
Viewed by 1111
Abstract
The use of supraphysiologic testosterone, particularly when alternated with an anti-androgen agent in men with metastatic castration-resistant prostate cancer (CRPC), has demonstrated promising results in clinical trials. As the use of this therapy in clinical practice is more widely adopted, there will be [...] Read more.
The use of supraphysiologic testosterone, particularly when alternated with an anti-androgen agent in men with metastatic castration-resistant prostate cancer (CRPC), has demonstrated promising results in clinical trials. As the use of this therapy in clinical practice is more widely adopted, there will be a growing need to understand the mechanisms of resistance. To that end, we independently derived three separate cell models of testosterone-sensitive CRPC. From each CRPC line, high dose testosterone-resistance (HTR) lines were selected. We demonstrated the differential response of the three CRPC lines to a high dose of testosterone in vitro and in vivo. We subsequently demonstrated the resistance of the HTR lines to testosterone and varying responses to testosterone withdrawal in vivo. The heterogeneity in responses to hormonal manipulation is correlated with varying levels of androgen receptor expression within the population. Overall, we show that we have developed three models of HTR that can be used to study the mechanisms of high dose testosterone resistance and identify potential therapeutic targets. Full article
Show Figures

Figure 1

Back to TopTop