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
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,595)

Search Parameters:
Keywords = prostate tumor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 19662 KiB  
Review
Partial Cystectomy for Muscle-Invasive Bladder Cancer
by Peter S. Palencia, Nethusan Sivanesan, Syed Rahman, Fady Ghali, David Hesse, John Colberg, Ashwin Sridhar, John D. Kelly, Byron H. Lee, Ashish M. Kamat and Wei-Shen Tan
Cancers 2025, 17(15), 2562; https://doi.org/10.3390/cancers17152562 (registering DOI) - 3 Aug 2025
Abstract
Partial cystectomy is a surgical bladder-sparing option for selected patients with muscle-invasive bladder cancer (MIBC), urachal adenocarcinoma and diverticular bladder tumors. Partial cystectomy hold several advantages. It allows for definite pathology and accurate staging while avoiding side effects from radiation therapy and preserves [...] Read more.
Partial cystectomy is a surgical bladder-sparing option for selected patients with muscle-invasive bladder cancer (MIBC), urachal adenocarcinoma and diverticular bladder tumors. Partial cystectomy hold several advantages. It allows for definite pathology and accurate staging while avoiding side effects from radiation therapy and preserves the option for salvage radical therapy (radical cystectomy or radical radiotherapy). Patients should have a CT urogram, prostatic urethral biopsy and mapping biopsies or blue light cystoscopy to rule out multifocal disease or CIS. Small solitary MIBC patients without carcinoma in situ in an area of the bladder where resection can be performed with negative margin would be the ideal candidates for partial cystectomy. Neoadjuvant systemic therapy is recommended for patients undergoing partial cystectomy. Partial cystectomy can be performed either by open or robotic approaches. When compared to radical cystectomy, partial cystectomy affords a lower complication rate and length of stay and better quality of life. Recurrence-free survival, cancer-specific survival and overall survival at 5 years is 39–67%, 62–84% and 45–70%, respectively. Following partial cystectomy, patients should have three monthly cystoscopy and urinary cytology for the first 24 months followed by 6-monthly cystoscopy for year 3 and 4 and then yearly for life. Cross-sectional imaging should be performed every 3–6 months for the first 2–3 years and then annually for 5 years. Full article
Show Figures

Figure 1

39 pages, 2336 KiB  
Review
Omics-Mediated Treatment for Advanced Prostate Cancer: Moving Towards Precision Oncology
by Yasra Fatima, Kirubel Nigusu Jobre, Enrique Gomez-Gomez, Bartosz Małkiewicz, Antonia Vlahou, Marika Mokou, Harald Mischak, Maria Frantzi and Vera Jankowski
Int. J. Mol. Sci. 2025, 26(15), 7475; https://doi.org/10.3390/ijms26157475 (registering DOI) - 2 Aug 2025
Abstract
Prostate cancer accounts for approximately 1.5 million new diagnoses and 400,000 deaths every year worldwide, and demographic projections indicate a near-doubling of both figures by 2040. Despite existing treatments, 10–20% of patients eventually progress to metastatic castration-resistant disease (mCRPC). The median overall survival [...] Read more.
Prostate cancer accounts for approximately 1.5 million new diagnoses and 400,000 deaths every year worldwide, and demographic projections indicate a near-doubling of both figures by 2040. Despite existing treatments, 10–20% of patients eventually progress to metastatic castration-resistant disease (mCRPC). The median overall survival (OS) after progression to mCPRC drops to 24 months, and efficacy drops severely after each additional line of treatment. Omics platforms have reached advanced levels and enable the acquisition of high-resolution large datasets that can provide insights into the molecular mechanisms underlying PCa pathology. Genomics, especially DDR (DNA damage response) gene alterations, detected via tissue and/or circulating tumor DNA, efficiently guides therapy in advanced prostate cancer. Given recent developments, we have performed a comprehensive literature search to cover recent research and clinical trial reports (over the last five years) that integrate omics along three converging trajectories in therapeutic development: (i) predicting response to approved agents with demonstrated survival benefits, (ii) stratifying patients to receive therapies in clinical trials, (iii) guiding drug development as part of drug repurposing frameworks. Collectively, this review is intended to serve as a comprehensive resource of recent advancements in omics-guided therapies for advanced prostate cancer, a clinical setting with existing clinical needs and poor outcomes. Full article
(This article belongs to the Special Issue Molecular Research on Prostate Cancer)
Show Figures

Figure 1

21 pages, 1133 KiB  
Review
Beyond Docetaxel: Targeting Resistance Pathways in Prostate Cancer Treatment
by Tayo Alex Adekiya
BioChem 2025, 5(3), 24; https://doi.org/10.3390/biochem5030024 - 1 Aug 2025
Viewed by 46
Abstract
Prostate cancer continues to be the most common cause of cancer-related disease and mortality among men worldwide, especially in the advanced stages, notably metastatic castration-resistant prostate cancer (mCRPC), which poses significant treatment challenges. Docetaxel, a widely used chemotherapeutic agent, has long served as [...] Read more.
Prostate cancer continues to be the most common cause of cancer-related disease and mortality among men worldwide, especially in the advanced stages, notably metastatic castration-resistant prostate cancer (mCRPC), which poses significant treatment challenges. Docetaxel, a widely used chemotherapeutic agent, has long served as the standard treatment, offering survival benefits and mitigation. However, its clinical impact is frequently undermined by the development of chemoresistance, which is a formidable challenge that leads to treatment failure and disease progression. The mechanisms driving docetaxel resistance are diverse and complex, encompassing modifications in androgen receptor signaling, drug efflux transporters, epithelial-mesenchymal transition (EMT), microtubule alterations, apoptotic pathway deregulation, and tumor microenvironmental influences. Recent evidence suggests that extracellular RNAs influence drug responses, further complicating the resistance landscape. This review offers a broad discussion on the mechanisms of resistance and explores novel therapeutic approaches to address them. These include next-generation taxanes, targeted molecular inhibitors, immunotherapies, and combination regimens that can be designed to counteract specific resistance pathways. By broadening our understanding of docetaxel resistance, this review highlights potential strategies to improve therapeutic efficacy and the potential to enhance outcomes in patients with advanced treatment-resistant prostate cancer. Full article
Show Figures

Figure 1

29 pages, 959 KiB  
Review
Machine Learning-Driven Insights in Cancer Metabolomics: From Subtyping to Biomarker Discovery and Prognostic Modeling
by Amr Elguoshy, Hend Zedan and Suguru Saito
Metabolites 2025, 15(8), 514; https://doi.org/10.3390/metabo15080514 (registering DOI) - 1 Aug 2025
Viewed by 122
Abstract
Cancer metabolic reprogramming plays a critical role in tumor progression and therapeutic resistance, underscoring the need for advanced analytical strategies. Metabolomics, leveraging mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy, offers a comprehensive and functional readout of tumor biochemistry. By enabling both targeted [...] Read more.
Cancer metabolic reprogramming plays a critical role in tumor progression and therapeutic resistance, underscoring the need for advanced analytical strategies. Metabolomics, leveraging mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy, offers a comprehensive and functional readout of tumor biochemistry. By enabling both targeted metabolite quantification and untargeted profiling, metabolomics captures the dynamic metabolic alterations associated with cancer. The integration of metabolomics with machine learning (ML) approaches further enhances the interpretation of these complex, high-dimensional datasets, providing powerful insights into cancer biology from biomarker discovery to therapeutic targeting. This review systematically examines the transformative role of ML in cancer metabolomics. We discuss how various ML methodologies—including supervised algorithms (e.g., Support Vector Machine, Random Forest), unsupervised techniques (e.g., Principal Component Analysis, t-SNE), and deep learning frameworks—are advancing cancer research. Specifically, we highlight three major applications of ML–metabolomics integration: (1) cancer subtyping, exemplified by the use of Similarity Network Fusion (SNF) and LASSO regression to classify triple-negative breast cancer into subtypes with distinct survival outcomes; (2) biomarker discovery, where Random Forest and Partial Least Squares Discriminant Analysis (PLS-DA) models have achieved >90% accuracy in detecting breast and colorectal cancers through biofluid metabolomics; and (3) prognostic modeling, demonstrated by the identification of race-specific metabolic signatures in breast cancer and the prediction of clinical outcomes in lung and ovarian cancers. Beyond these areas, we explore applications across prostate, thyroid, and pancreatic cancers, where ML-driven metabolomics is contributing to earlier detection, improved risk stratification, and personalized treatment planning. We also address critical challenges, including issues of data quality (e.g., batch effects, missing values), model interpretability, and barriers to clinical translation. Emerging solutions, such as explainable artificial intelligence (XAI) approaches and standardized multi-omics integration pipelines, are discussed as pathways to overcome these hurdles. By synthesizing recent advances, this review illustrates how ML-enhanced metabolomics bridges the gap between fundamental cancer metabolism research and clinical application, offering new avenues for precision oncology through improved diagnosis, prognosis, and tailored therapeutic strategies. Full article
(This article belongs to the Special Issue Nutritional Metabolomics in Cancer)
Show Figures

Figure 1

12 pages, 1472 KiB  
Article
Furosemide Reduces Radionuclide Activity in the Bladder in 18F-PSMA-1007-PET/CT: A Single-Center Retrospective Intra-Individual Comparative Study
by Martin A. Cahenzli, Andreas S. Kreusch, Philipp Huber, Marco Dressler, Janusch P. Blautzik and Gregor Sommer
Diagnostics 2025, 15(15), 1931; https://doi.org/10.3390/diagnostics15151931 - 31 Jul 2025
Viewed by 90
Abstract
Background/Objectives: 18F-PSMA-1007 is one of the more widely used radioligands in prostate cancer imaging with PET/CT. Its major advantage lies in the low urinary tracer activity due to primarily hepatobiliary clearance, but unexpectedly high tracer accumulation in the bladder can occur, [...] Read more.
Background/Objectives: 18F-PSMA-1007 is one of the more widely used radioligands in prostate cancer imaging with PET/CT. Its major advantage lies in the low urinary tracer activity due to primarily hepatobiliary clearance, but unexpectedly high tracer accumulation in the bladder can occur, potentially hindering assessment of lesions near the prostate bed. This study assesses the impact of furosemide on 18F-PSMA-1007 tracer accumulation in the bladder. Methods: In this single-center, retrospective, intra-individual comparative analysis, 18 patients undergoing two consecutive 18F-PSMA-1007 PET/CT scans for biochemical relapse (BCR) or persistence (BCP)—one with and one without prior furosemide administration—were included. Images were acquired 60 min post-injection of 250 MBq of tracer activity. Standardized Uptake Values (SUVmax, SUVpeak, SUVmean) were measured in the bladder and in tissues with physiological uptake by three readers. Differences were analyzed using Wilcoxon signed-rank tests. The inter-reader agreement was assessed using intraclass correlation coefficient. Results: Furosemide significantly decreased bladder SUVmax, SUVpeak, and SUVmean (all p < 0.001). Mean bladder SUVmax decreased from 13.20 ± 10.40 to 3.92 ± 3.47, SUVpeak from 10.94 ± 8.02 to 3.47 ± 3.13, and SUVmean from 8.74 ± 6.66 to 2.81 ± 2.56, representing a large effect size (r ≈ 0.55). Physiological tracer uptake in most organs was not significantly influenced by furosemide (all p > 0.05). Conclusions: Despite the predominantly hepatobiliary clearance of 18F-PSMA-1007, furosemide-induced forced diuresis leads to a significant reduction in tracer activity in the bladder, which in clinical practice could help in early detection of tumor recurrence. Full article
(This article belongs to the Special Issue Research Update on Nuclear Medicine)
Show Figures

Figure 1

15 pages, 281 KiB  
Article
Single-Center Comparative Evaluation of Freehand Transperineal and Transrectal Prostate Biopsy Techniques Performed Under Local Anesthesia
by Laurian Ștefan Maxim, Ruxandra Maria Rotaru, Camelia Cornelia Scârneciu, Marius Alexandru Moga, Florin Lucian Petrică Sabou, Anda Catica Hogea, Raul Dumitru Gherasim, Alexandru Ghicavîi, Razvan-Dragos Mulțescu, Mihail-Alexandru Badea, Bogdan Ovidiu Feciche and Ioan Scârneciu
Diagnostics 2025, 15(15), 1929; https://doi.org/10.3390/diagnostics15151929 - 31 Jul 2025
Viewed by 169
Abstract
Background: To diagnose prostate cancer, a prostate biopsy is required. Two methods are commonly used for biopsy: transrectal and transperineal. The transperineal approach, particularly the “freehand” technique under local anesthesia, offers better access to the anterior prostate, lower infection risk, and higher detection [...] Read more.
Background: To diagnose prostate cancer, a prostate biopsy is required. Two methods are commonly used for biopsy: transrectal and transperineal. The transperineal approach, particularly the “freehand” technique under local anesthesia, offers better access to the anterior prostate, lower infection risk, and higher detection rates. Methods: This retrospective study examined the records of 1674 patients who underwent ultrasound-guided prostate biopsies between 2015 and 2022. Of these, 1161 patients had transperineal biopsy using the “freehand” method, and 513 had transrectal biopsy. All the biopsies were carried out under local anesthesia, with a combined systematic and targeted approach for patients with MRI-identified lesions. Results: This study demonstrates that the transperineal biopsy approach significantly increased the detection rate of clinically significant prostate cancer compared with the transrectal method, with detection rates of 65.7% and 59.4%, respectively. Notably, the transperineal technique also achieved superior detection of anteriorly located tumors (94.1% vs. 43.1%), supporting its use as the preferred biopsy strategy, particularly in anatomically challenging regions. Moreover, patients who underwent transperineal biopsy demonstrated more favorable diagnostic outcomes, characterized by a higher detection rate for clinically significant cancers and a reduced incidence of clinically insignificant cases. The transperineal method outperformed the transrectal approach, especially among younger patients and those presenting with lower PSA values. These results highlight the diagnostic superiority and broader clinical applicability of the transperineal biopsy technique across various patient subgroups. Full article
(This article belongs to the Special Issue Recent Advances in Prostate Cancer Imaging and Biopsy Techniques)
15 pages, 2220 KiB  
Article
Radiologic Assessment of Periprostatic Fat as an Indicator of Prostate Cancer Risk on Multiparametric MRI
by Roxana Iacob, Emil Radu Iacob, Emil Robert Stoicescu, Diana Manolescu, Laura Andreea Ghenciu, Radu Căprariu, Amalia Constantinescu, Iulia Ciobanu, Răzvan Bardan and Alin Cumpănaș
Bioengineering 2025, 12(8), 831; https://doi.org/10.3390/bioengineering12080831 (registering DOI) - 31 Jul 2025
Viewed by 149
Abstract
Prostate cancer remains one of the most prevalent malignancies among men, and emerging evidence proposed a potential role for periprostatic adipose tissue (PPAT) in tumor progression. However, its relationship with imaging-based risk stratification systems such as PI-RADS remains uncertain. This retrospective observational study [...] Read more.
Prostate cancer remains one of the most prevalent malignancies among men, and emerging evidence proposed a potential role for periprostatic adipose tissue (PPAT) in tumor progression. However, its relationship with imaging-based risk stratification systems such as PI-RADS remains uncertain. This retrospective observational study aimed to evaluate whether periprostatic and subcutaneous fat thickness are associated with PI-RADS scores or PSA levels in biopsy-naïve patients. We retrospectively reviewed 104 prostate MRI scans performed between January 2020 and January 2024. Fat thickness was measured on axial T2-weighted images, and statistical analyses were conducted using Spearman’s correlation and multiple linear regression. In addition to linear measurements, we also assessed periprostatic fat volume and posterior fat thickness derived from imaging data. No significant correlations were observed between fat thickness (either periprostatic or subcutaneous) and PI-RADS score or PSA values. Similarly, periprostatic fat volume showed only a weak, non-significant correlation with PI-RADS, while posterior fat thickness demonstrated a weak but statistically significant positive association. Additionally, subgroup comparisons between low-risk (PI-RADS < 4) and high-risk (PI-RADS ≥ 4) patients showed no meaningful differences in fat measurements. These findings suggest that simple linear fat thickness measurements may not enhance imaging-based risk assessment in prostate cancer, though regional and volumetric assessments could offer modest added value. Full article
(This article belongs to the Special Issue Label-Free Cancer Detection)
Show Figures

Figure 1

38 pages, 2158 KiB  
Review
Epigenetic Modulation and Bone Metastasis: Evolving Therapeutic Strategies
by Mahmoud Zhra, Jasmine Hanafy Holail and Khalid S. Mohammad
Pharmaceuticals 2025, 18(8), 1140; https://doi.org/10.3390/ph18081140 - 31 Jul 2025
Viewed by 349
Abstract
Bone metastasis remains a significant cause of morbidity and diminished quality of life in patients with advanced breast, prostate, and lung cancers. Emerging research highlights the pivotal role of reversible epigenetic alterations, including DNA methylation, histone modifications, chromatin remodeling complex dysregulation, and non-coding [...] Read more.
Bone metastasis remains a significant cause of morbidity and diminished quality of life in patients with advanced breast, prostate, and lung cancers. Emerging research highlights the pivotal role of reversible epigenetic alterations, including DNA methylation, histone modifications, chromatin remodeling complex dysregulation, and non-coding RNA networks, in orchestrating each phase of skeletal colonization. Site-specific promoter hypermethylation of tumor suppressor genes such as HIN-1 and RASSF1A, alongside global DNA hypomethylation that activates metastasis-associated genes, contributes to cancer cell plasticity and facilitates epithelial-to-mesenchymal transition (EMT). Key histone modifiers, including KLF5, EZH2, and the demethylases KDM4/6, regulate osteoclastogenic signaling pathways and the transition between metastatic dormancy and reactivation. Simultaneously, SWI/SNF chromatin remodelers such as BRG1 and BRM reconfigure enhancer–promoter interactions that promote bone tropism. Non-coding RNAs, including miRNAs, lncRNAs, and circRNAs (e.g., miR-34a, NORAD, circIKBKB), circulate via exosomes to modulate the RANKL/OPG axis, thereby conditioning the bone microenvironment and fostering the formation of a pre-metastatic niche. These mechanistic insights have accelerated the development of epigenetic therapies. DNA methyltransferase inhibitors (e.g., decitabine, guadecitabine) have shown promise in attenuating osteoclast differentiation, while histone deacetylase inhibitors display context-dependent effects on tumor progression and bone remodeling. Inhibitors targeting EZH2, BET proteins, and KDM1A are now advancing through early-phase clinical trials, often in combination with bisphosphonates or immune checkpoint inhibitors. Moreover, novel approaches such as CRISPR/dCas9-based epigenome editing and RNA-targeted therapies offer locus-specific reprogramming potential. Together, these advances position epigenetic modulation as a promising axis in precision oncology aimed at interrupting the pathological crosstalk between tumor cells and the bone microenvironment. This review synthesizes current mechanistic understanding, evaluates the therapeutic landscape, and outlines the translational challenges ahead in leveraging epigenetic science to prevent and treat bone metastases. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Graphical abstract

20 pages, 2717 KiB  
Article
Unlocking the Potential of Gracilaria chilensis Against Prostate Cancer
by Verónica Torres-Estay, Lorena Azocar, Camila Schmidt, Macarena Aguilera-Olguín, Catalina Ramírez-Santelices, Emilia Flores-Faúndez, Paula Sotomayor, Nancy Solis, Daniel Cabrera, Loretto Contreras-Porcia, Francisca C. Bronfman and Alejandro S. Godoy
Plants 2025, 14(15), 2352; https://doi.org/10.3390/plants14152352 - 31 Jul 2025
Viewed by 231
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death among men in most Western countries. Current therapies for PCa are limited, often ineffective, and associated with significant side effects. As a result, there is a growing interest in exploring new therapeutic [...] Read more.
Prostate cancer (PCa) is the second leading cause of cancer-related death among men in most Western countries. Current therapies for PCa are limited, often ineffective, and associated with significant side effects. As a result, there is a growing interest in exploring new therapeutic agents, particularly from the polyphyletic group of algae, which offers a promising source of compounds with anticancer properties. Our research group has focused on investigating the effects of a novel oleoresin from Gracilaria chilensis, known as Gracilex®, as a potential therapeutic agent against PCa using both in vitro and in vivo models. Our findings indicate that Gracilex® exhibits a time- and dose-dependent inhibitory effect on cell survival in LNCaP and PC-3 PCa, reducing viability by over 50% and inducing apoptosis, as evidenced by a significant increase in activated caspase-3 expression in both cell lines. Moreover, Gracilex® significantly reduces the proliferation rate of both LNCaP and PC-3 prostate cancer cell lines, as evidenced by a marked decrease in the growth curve slope (p = 0.0034 for LNCaP; p < 0.0001 for PC-3) and a 40–50% reduction in the proportion of Ki-67-positive PCa cells. In addition, Gracilex® significantly reduces in vitro cell migration and invasion in LNCaP and PC-3 cell lines. Lastly, Gracilex® inhibits tumor growth in an in vivo xenograft model, an effect that correlates with the reduced PCa cell proliferation observed in tumor tissue sections. Collectively, our data strongly support the broad antitumoral effects of Gracilex® on PCa cells in vitro and in vivo. These findings advance our understanding of its potential therapeutic role in PCa and highlight the relevance of further investigating algae-derived compounds for cancer treatment. Full article
(This article belongs to the Section Plant Genetics, Genomics and Biotechnology)
Show Figures

Figure 1

21 pages, 2038 KiB  
Article
Germline BARD1 Mutation in High-Risk Chinese Breast and Ovarian Cancer Patients
by Ava Kwong, Cecilia Y. S. Ho, Chun Hang Au and Edmond S. K. Ma
Cancers 2025, 17(15), 2524; https://doi.org/10.3390/cancers17152524 - 30 Jul 2025
Viewed by 187
Abstract
Background: The prevalence of BARD1 mutations in breast and ovarian cancers varies across different ethnic groups. Evaluating the cancer risk and clinical significance of BARD1 mutations in the local Chinese patients with breast cancer, ovarian cancer, or both is clinically important for designing [...] Read more.
Background: The prevalence of BARD1 mutations in breast and ovarian cancers varies across different ethnic groups. Evaluating the cancer risk and clinical significance of BARD1 mutations in the local Chinese patients with breast cancer, ovarian cancer, or both is clinically important for designing an appropriate surveillance scheme. Methods: This study used a 30 gene panel to identify BARD1 germline mutations in 2658 breast and ovarian cancer patients. Results: Among this cohort, the BARD1 mutation prevalence was 0.45% for breast cancer and 0.29% for ovarian cancer. In our 12 mutation carriers, we identified eight types of mutation variants, including three novel mutations. BARD1 mutation carriers were more likely to have a family history of liver, prostate, and cervical cancers (p-values = 0.004, 0.018, and 0.037, respectively) than patients who tested negative for mutations. Among the BARD1 mutants, the majority of the breast tumors were invasive ductal carcinoma (NOS type) (10/11, 90.9%) of high-grade disease (9/9, 100%) and half of them were triple-negative breast cancer (5/10, 50%). Conclusions: Although the prevalence of BARD1 mutations is low and the penetrance is incomplete, we recommend including BARD1 in the test panel for breast cancer patients. Our data suggest that more comprehensive surveillance management may be considered in mutation carriers due to the familial aggregation of a relatively wide spectrum of cancers. Full article
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)
Show Figures

Figure 1

17 pages, 481 KiB  
Review
Cognitive Impairment in Prostate Cancer Patients Receiving Androgen Deprivation Therapy: A Scoping Review
by João Vasco Barreira, Pedro Barreira, Gil Falcão, Daniela Garcez, Pedro Silva, Gustavo Santos, Mário Fontes-Sousa, José Leão Mendes, Filipa Reis, Carla F. Santos, Filipa Ribeiro and Manuel Luís Capelas
Cancers 2025, 17(15), 2501; https://doi.org/10.3390/cancers17152501 - 29 Jul 2025
Viewed by 214
Abstract
Background: Androgen deprivation therapy (ADT) is a primary treatment for prostate cancer (PCa) that effectively reduces androgen levels to suppress tumor progression. However, growing evidence suggests potential cognitive side effects, raising concerns about the long-term neurological consequences of this treatment. Objective: This scoping [...] Read more.
Background: Androgen deprivation therapy (ADT) is a primary treatment for prostate cancer (PCa) that effectively reduces androgen levels to suppress tumor progression. However, growing evidence suggests potential cognitive side effects, raising concerns about the long-term neurological consequences of this treatment. Objective: This scoping review aims to synthesize the existing evidence linking ADT to cognitive changes in men with PCa, identifying the key cognitive domains affected and outlining gaps in the existing literature. Methods: A systematic literature search was conducted according to the PRISMA-ScR guidelines in CINAHL, PubMed, Scopus, and Web of Science. Studies investigating cognitive function in ADT-treated PCa patients were included, covering randomized controlled trials (RCTs) and cohort, case–control, and cross-sectional studies. The extracted data included the study design, evaluated cognitive characteristics, measurement tools, and overall findings. Results: A total of 22 studies met the inclusion and exclusion criteria. Cognitive assessments varied across studies. While some studies reported cognitive impairments in ADT-treated patients—particularly in working, verbal, and visual memory and executive function—others found no significant effects. The variability in prostate cancer staging, epidemiological study designs, and treatment regimens; the exclusion of comorbid conditions; and the differences in assessment tools, sample sizes, and study durations hinder definitive conclusions about the cognitive effects of ADT. Conclusions: This scoping review highlights the heterogeneous and often contradictory evidence regarding ADT-associated cognitive dysfunction. While certain cognitive domains may be affected, methodological inconsistencies limit robust conclusions. Standardized cognitive assessments and longer longitudinal studies are required to clarify ADT’s role in cognitive decline. As the PCa survival rate increases with extended ADT use, integrating routine cognitive monitoring into clinical practice should be considered for PCa patients. Full article
(This article belongs to the Special Issue Novel Insights into Cancer-Related Cognitive Impairment)
Show Figures

Figure 1

26 pages, 1745 KiB  
Review
Emerging PET Imaging Agents and Targeted Radioligand Therapy: A Review of Clinical Applications and Trials
by Maierdan Palihati, Jeeban Paul Das, Randy Yeh and Kathleen Capaccione
Tomography 2025, 11(8), 83; https://doi.org/10.3390/tomography11080083 - 28 Jul 2025
Viewed by 423
Abstract
Targeted radioligand therapy (RLT) is an emerging field in anticancer therapeutics with great potential across tumor types and stages of disease. While much progress has focused on agents targeting somatostatin receptors and prostate-specific membrane antigen (PSMA), the same advanced radioconjugation methods and molecular [...] Read more.
Targeted radioligand therapy (RLT) is an emerging field in anticancer therapeutics with great potential across tumor types and stages of disease. While much progress has focused on agents targeting somatostatin receptors and prostate-specific membrane antigen (PSMA), the same advanced radioconjugation methods and molecular targeting have spurred the development of numerous theranostic combinations for other targets. A number of the most promising agents have progressed to clinical trials and are poised to change the landscape of positron emission tomography (PET) imaging. Here, we present recent data on some of the most important emerging molecular targeted agents with their exemplar clinical images, including agents targeting fibroblast activation protein (FAP), hypoxia markers, gastrin-releasing peptide receptors (GRPrs), and integrins. These radiopharmaceuticals share the promising characteristic of being able to image multiple types of cancer. Early clinical trials have already demonstrated superiority to 18F-fluorodeoxyglucose (18F-FDG) for some, suggesting the potential to supplant this longstanding PET radiotracer. Here, we provide a primer for practicing radiologists, particularly nuclear medicine clinicians, to understand novel PET imaging agents and their clinical applications, as well as the availability of companion targeted radiotherapeutics, the status of their regulatory approval, the potential challenges associated with their use, and the future opportunities and perspectives. Full article
(This article belongs to the Section Cancer Imaging)
Show Figures

Figure 1

26 pages, 2998 KiB  
Review
PSMA-Directed Theranostics in Prostate Cancer
by Salman Ayub Jajja, Nandini Sodhi, Ephraim E. Parent and Parminder Singh
Biomedicines 2025, 13(8), 1837; https://doi.org/10.3390/biomedicines13081837 - 28 Jul 2025
Viewed by 727
Abstract
Following lung cancer, prostate cancer is the leading cause of cancer death in men. High-risk localized tumor burden or metastatic disease often progresses, refractory to initial treatment regimens. There is ongoing development of technology to appropriately identify high-risk patients, stage them correctly, and [...] Read more.
Following lung cancer, prostate cancer is the leading cause of cancer death in men. High-risk localized tumor burden or metastatic disease often progresses, refractory to initial treatment regimens. There is ongoing development of technology to appropriately identify high-risk patients, stage them correctly, and offer appropriate treatments to obtain the best clinical outcomes. Prostate cancer-specific membrane antigen (PSMA) is a transmembrane glutamate carboxypeptidase, which helps regulate folate absorption, and its overexpression is pathologically directly proportional and associated with prostate cancer. Increased PSMA expression is a known independent risk factor for poorer survival, and most metastatic lesions in CRPC are PSMA positive. Over the last decade, several PSMA-based PET radiopharmaceuticals have demonstrated superior sensitivities and specificities compared to traditional imaging methods. These outcomes have been demonstrated by several large clinical trials. As the data emerges, these diagnostics are being integrated into standard of care protocol to facilitate nuanced identification of malignant lesions. PSMA is also being targeted through several therapeutics, including radioligands and immunotherapies such as CAR-T, BiTEs, and ADCs. This review will discuss the landscape of PSMA-based theranostics in the context of prostate cancer. Full article
(This article belongs to the Special Issue Advanced Research on Genitourinary Cancer)
Show Figures

Figure 1

37 pages, 3106 KiB  
Review
Quantum Dot-Enabled Biosensing for Prostate Cancer Diagnostics
by Hossein Omidian, Erma J. Gill and Luigi X. Cubeddu
Nanomaterials 2025, 15(15), 1162; https://doi.org/10.3390/nano15151162 - 28 Jul 2025
Viewed by 234
Abstract
Prostate cancer diagnostics are rapidly advancing through innovations in nanotechnology, biosensing strategies, and molecular recognition. This review analyzes studies focusing on quantum dot (QD)-based biosensors for detecting prostate cancer biomarkers with high sensitivity and specificity. It covers diverse sensing platforms and signal transduction [...] Read more.
Prostate cancer diagnostics are rapidly advancing through innovations in nanotechnology, biosensing strategies, and molecular recognition. This review analyzes studies focusing on quantum dot (QD)-based biosensors for detecting prostate cancer biomarkers with high sensitivity and specificity. It covers diverse sensing platforms and signal transduction mechanisms, emphasizing the influence of the QD composition, surface functionalization, and bio interface engineering on analytical performance. Key metrics such as detection limits, dynamic range, and compatibility with biological samples, including serum, urine, and tissue, are critically assessed. Recent advances in green-synthesized QDs and smartphone-integrated diagnostic platforms are highlighted, including lateral flow assays, paper-based devices, and pH-responsive hydrogels for real-time, low-cost, and decentralized cancer screening. These innovations enable multiplexed biomarker detection and tumor microenvironment monitoring in point-of-care settings. This review concludes by addressing the current limitations, scalability challenges, and future research directions for translating QD-enabled biosensors into clinically viable diagnostic tools. Full article
(This article belongs to the Section Nanofabrication and Nanomanufacturing)
Show Figures

Figure 1

16 pages, 5172 KiB  
Article
LAMP1 as a Target for PET Imaging in Adenocarcinoma Xenograft Models
by Bahar Ataeinia, Arvin Haj-Mirzaian, Lital Ben-Naim, Shadi A. Esfahani, Asier Marcos Vidal, Umar Mahmood and Pedram Heidari
Pharmaceuticals 2025, 18(8), 1122; https://doi.org/10.3390/ph18081122 - 27 Jul 2025
Viewed by 432
Abstract
Background: Lysosomal-associated membrane protein 1 (LAMP1), typically localized to the lysosomal membrane, is increasingly implicated as a marker of cancer aggressiveness and metastasis when expressed on the cell surface. This study aimed to develop a LAMP1-targeted antibody-based PET tracer and assess its efficacy [...] Read more.
Background: Lysosomal-associated membrane protein 1 (LAMP1), typically localized to the lysosomal membrane, is increasingly implicated as a marker of cancer aggressiveness and metastasis when expressed on the cell surface. This study aimed to develop a LAMP1-targeted antibody-based PET tracer and assess its efficacy in mouse models of human breast and colon adenocarcinoma. Methods: To determine the source of LAMP1 expression, we utilized human single-cell RNA sequencing and spatial transcriptomics, complemented by in-house flow cytometry on xenografted mouse models. Tissue microarrays of multiple epithelial cancers and normal tissue were stained for LAMP-1, and staining was quantified. An anti-LAMP1 monoclonal antibody was conjugated with desferrioxamine (DFO) and labeled with zirconium-89 (89Zr). Human triple-negative breast cancer (MDA-MB-231) and colon cancer (Caco-2) cell lines were implanted in nude mice. PET/CT imaging was conducted at 24, 72, and 168 h post-intravenous injection of 89Zr-DFO-anti-LAMP1 and 89Zr-DFO-IgG (negative control), followed by organ-specific biodistribution analyses at the final imaging time point. Results: Integrated single-cell and spatial RNA sequencing demonstrated that LAMP1 expression was localized to myeloid-derived suppressor cells (MDSCs) and cancer-associated fibroblasts (CAFs) in addition to the cancer cells. Tissue microarray showed significantly higher staining for LAMP-1 in tumor tissue compared to normal tissue (3986 ± 2635 vs. 1299 ± 1291, p < 0.001). Additionally, xenograft models showed a significantly higher contribution of cancer cells than the immune cells to cell surface LAMP1 expression. In vivo, PET imaging with 89Zr-DFO-anti-LAMP1 PET/CT revealed detectable tumor uptake as early as 24 h post-injection. The 89Zr-DFO-anti-LAMP1 tracer demonstrated significantly higher uptake than the control 89Zr-DFO-IgG in both models across all time points (MDA-MB-231 SUVmax at 168 h: 12.9 ± 5.7 vs. 4.4 ± 2.4, p = 0.003; Caco-2 SUVmax at 168 h: 8.53 ± 3.03 vs. 3.38 ± 1.25, p < 0.01). Conclusions: Imaging of cell surface LAMP-1 in breast and colon adenocarcinoma is feasible by immuno-PET. LAMP-1 imaging can be expanded to adenocarcinomas of other origins, such as prostate and pancreas. Full article
Show Figures

Figure 1

Back to TopTop