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
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
remove_circle_outline

Search Results (3,614)

Search Parameters:
Keywords = prostate cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 3069 KB  
Article
Cyclophilin Inhibitor Rencofilstat Combined with Proteasome Inhibitor Ixazomib Increases Proteotoxic Cell Death in Advanced Prostate Cancer Cells with Minimal Effects on Non-Cancer Cells
by Carlos Perez-Stable, Alicia de las Pozas, Medhi Wangpaichitr, Robert T. Foster and Daren R. Ure
Biomedicines 2025, 13(10), 2442; https://doi.org/10.3390/biomedicines13102442 (registering DOI) - 7 Oct 2025
Abstract
Background/Objective: Proteotoxic stress induced by inhibitors of the ubiquitin–proteasome system has been successful in multiple myeloma but not in solid cancers such as prostate cancer. Our objective is to find a combination with proteasome inhibitors that increases apoptotic cell death in all types [...] Read more.
Background/Objective: Proteotoxic stress induced by inhibitors of the ubiquitin–proteasome system has been successful in multiple myeloma but not in solid cancers such as prostate cancer. Our objective is to find a combination with proteasome inhibitors that increases apoptotic cell death in all types of prostate cancer without harming non-cancer cells. Methods: The effectiveness of rencofilstat, a pan-cyclophilin inhibitor, combined with the ixazomib proteasome inhibitor, was investigated in multiple prostate cancer and non-cancer cells. Inducible knockdown of stress response XBP1s and cyclophilins A/B and inducible expression of XBP1s and cyclophilin B were developed in prostate cancer to determine functional roles. Results: Rencofilstat + ixazomib increased apoptotic cell death in prostate cancer but not in non-cancer cells. We investigated the effects on XBP1s and PERK, important unfolded protein response factors required for cells to survive proteotoxic stress. The results revealed that XBP1s had a pro-survival role early, but maintenance at later times of rencofilstat + ixazomib treatment resulted in cell death. In addition, decreased PERK and phospho-eIF2α likely maintained protein synthesis to further enhance proteotoxic stress. In contrast, rencofilstat + ixazomib did not alter XBP1s or PERK in non-cancer cells. Additional genetic experiments showed that the RCF targets cyclophilins A, B, and D had protective effects. Rencofilstat increased extracellular secretion of cyclophilin B, but rencofilstat + ixazomib reduced glycosylation and, likely, the biological function of CD147 (CypB receptor) and decreased downstream ERK signaling. Conclusions: Rencofilstat + ixazomib may be a new strategy for increasing proteotoxic stress and apoptotic cell death in advanced prostate cancer cells with less toxic side effects. Full article
Show Figures

Figure 1

15 pages, 4716 KB  
Review
Coumarin–Dithiocarbamate Derivatives as Biological Agents
by Piotr Wiliński, Aleksander Kurzątkowski and Kinga Ostrowska
Int. J. Mol. Sci. 2025, 26(19), 9667; https://doi.org/10.3390/ijms26199667 - 3 Oct 2025
Abstract
Coumarin derivatives, whether natural or synthetic, have attracted considerable interest from medicinal chemists due to their versatile biological properties. Their appealing pharmacological activities—such as anticancer, anti-inflammatory, neuroprotective, anticoagulant, and antioxidant effects—combined with the ease of their synthesis and the ability to introduce chemical [...] Read more.
Coumarin derivatives, whether natural or synthetic, have attracted considerable interest from medicinal chemists due to their versatile biological properties. Their appealing pharmacological activities—such as anticancer, anti-inflammatory, neuroprotective, anticoagulant, and antioxidant effects—combined with the ease of their synthesis and the ability to introduce chemical modifications at multiple positions have made them a widely explored class of compounds. In the scientific literature, there are many examples. On the other hand, dithiocarbamates, originally employed as pesticides and fungicides in agriculture, have recently emerged as potential therapeutic agents for the treatment of serious diseases such as cancer and microbial infections. Moreover, dithiocarbamates bearing diverse organic functionalities have demonstrated significant antifungal properties against resistant phytopathogenic fungi, presenting a promising approach to combat the growing global issue of fungal resistance. Dithiocarbamates linked to coumarin derivatives have been shown to exhibit cytotoxic activity against various human cancer cell lines, including MGC-803 (gastric), MCF-7 (breast), PC-3 (prostate), EC-109 (esophageal), H460 (non-small cell lung), HCCLM-7 (hepatocellular carcinoma), HeLa (cervical carcinoma), MDA-MB-435S (mammary adenocarcinoma), SW480 (colon carcinoma), and Hep-2 (laryngeal carcinoma). Numerous studies have revealed that the inclusion of a dithiocarbamate moiety can provide central nervous system (CNS) activity, particularly through inhibitory potency and selectivity toward acetylcholinesterase (AChE) and monoamine oxidases (MAO-A and MAO-B). Recently, it has been reported that coumarin–dithiocarbamate derivatives exhibit α-glucosidase inhibitory effects and also possess promising antimicrobial activity. This study presents an overview of recent progress in the chemistry of coumarin–dithiocarbamate derivatives, with a focus on their biological activity. Previous review papers focused on coumarin derivatives as multitarget compounds for neurodegenerative diseases and described various types of compounds, with dithiocarbamate derivatives representing only a small part of them. Our work deals exclusively with coumarin dithiocarbamates and their biological activity. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

12 pages, 968 KB  
Article
Preclinical Theranostic Profiling of [64Cu]Cu-Acetate in Prostate Cancer
by Sadaf Ghanaatgar Kasbi, Martin Savard, Céléna Dubuc, Yves Dory, Brigitte Guérin and Fernand Gobeil
Molecules 2025, 30(19), 3957; https://doi.org/10.3390/molecules30193957 - 2 Oct 2025
Abstract
Copper plays a critical role in cancer biology, with tumor cells exhibiting abnormal copper metabolism that drives proliferation and tumor growth. A limited number of preclinical and clinical studies have reported promising theranostic potential of copper-based radionuclides, such as 64Cu, for both [...] Read more.
Copper plays a critical role in cancer biology, with tumor cells exhibiting abnormal copper metabolism that drives proliferation and tumor growth. A limited number of preclinical and clinical studies have reported promising theranostic potential of copper-based radionuclides, such as 64Cu, for both diagnostic imaging and targeted radiotherapy in diverse cancers, including prostate cancer (PCa). In this work, we evaluated the cellular uptake and antitumor efficacy of [64Cu]Cu-acetate using both cellular and animal models of PCa. Uptake assays revealed that ~70% of the administered dose (10 kBq) was internalized by PC-3 cells within 24 h, predominantly localizing to the cytoplasm, with around 9% detected in the nucleus. These results were corroborated by comparable natural Cu-acetate uptake levels (at equimolar dose) in PC-3 cells, as quantified by ICP-MS. Clonogenic assays revealed a dose-dependent reduction in survival following treatment with [64Cu]Cu-acetate (3 and 6 MBq), whereas its non-radioactive counterpart [NatCu]Cu-acetate, even at excess concentrations (10 µM), had no significant effect. Ex vivo biodistribution studies showed selective tumor accumulation/retention alongside expected hepatic uptake. Clear tumor visualization was achieved using μPET imaging with [64Cu]Cu-acetate (10 MBq iv). A single higher dose (65 MBq iv) effectively reduced tumor growth in a subcutaneous PC-3 xenograft mouse model, without systemic toxicity, as evidenced by stable body weight. Together, these results further support the theranostic potential of [64Cu]Cu in PCa. Full article
(This article belongs to the Special Issue Applications of Radiochemistry in Healthcare)
Show Figures

Figure 1

15 pages, 434 KB  
Review
Dendritic Cell Immunotherapy for Solid Tumors: Advances in Translational Research and Clinical Application
by Mi Eun Kim and Jun Sik Lee
Curr. Issues Mol. Biol. 2025, 47(10), 806; https://doi.org/10.3390/cimb47100806 - 1 Oct 2025
Abstract
Dendritic cells (DCs) are critical antigen-presenting cells that orchestrate the interface between innate and adaptive immunity, making them attractive approaches for cancer immunotherapy. Recent advances in the characterization of DC subsets, antigen delivery strategies, and adjuvant design have enabled the enhancement of DC-based [...] Read more.
Dendritic cells (DCs) are critical antigen-presenting cells that orchestrate the interface between innate and adaptive immunity, making them attractive approaches for cancer immunotherapy. Recent advances in the characterization of DC subsets, antigen delivery strategies, and adjuvant design have enabled the enhancement of DC-based vaccines for solid tumors. Clinical studies across melanoma, glioblastoma, prostate cancer, and non-small cell lung cancer have demonstrated safety and immunogenicity, with encouraging signals of clinical efficacy, particularly when DC vaccination is combined with immune checkpoint blockade or personalized neoantigen approaches. However, translational barriers remain, including the immunosuppressive tumor microenvironment, inefficient DC migration, and variability in manufacturing protocols. Developing solutions such as in vivo DC targeting, biomaterials-based delivery systems, high-resolution single-cell analyses, and artificial intelligence-driven epitope prediction are controlled to overcome these challenges. Together, these innovations highlight the evolving role of DC immunotherapy as a foundation of precision oncology, offering the potential to integrate personalized vaccination strategies into standard treatment paradigms for solid tumors. Therefore, in this review, we specifically focus on these advances in dendritic cell immunotherapy for solid tumors and their translational implications. Full article
(This article belongs to the Special Issue Future Challenges of Targeted Therapy of Cancers: 2nd Edition)
Show Figures

Figure 1

15 pages, 309 KB  
Case Report
Psychological Involvement in the Journey of a Patient with Localized Prostate Cancer—From Diagnosis to Treatment
by Daniela Mihalcia Ailene, Gabriela Rahnea-Nita, Alexandru Nechifor, Liliana Florina Andronache, Mihaela Emilia Dumitru, Alexandru-Mihai Rebegea, Cristina Stefanescu, Roxana-Andreea Rahnea-Nita and Laura-Florentina Rebegea
Diseases 2025, 13(10), 319; https://doi.org/10.3390/diseases13100319 - 1 Oct 2025
Abstract
Introduction: Prostate cancer is one of the most common neoplasia in men, and its clinical evolution is highly influenced by psycho-emotional factors, especially in elderly patients. Comorbidities, the perception of one’s identity and its impact on life quality become relevant variables in the [...] Read more.
Introduction: Prostate cancer is one of the most common neoplasia in men, and its clinical evolution is highly influenced by psycho-emotional factors, especially in elderly patients. Comorbidities, the perception of one’s identity and its impact on life quality become relevant variables in the therapeutic decision. Sexual dysfunction after treatment along with decreased libido, erectile dysfunction and ejaculatory dysfunction are significant problems in patients with prostate cancer. Case presentation: The present study presents the oncological evolution of an elderly patient with a dual diagnosis, prostate adenocarcinoma and lung squamous cell carcinoma, who faced a significant amount of medical and psychological challenges. Reluctance to hormone therapy was closely linked to the fear of sexual dysfunction, a very common reaction in elderly men concerned with maintaining autonomy and intimacy. The peculiarity of the case consists in the interaction between the evolution of the disease, the therapeutic decisions and the psychological impact on the patient. Discussion: Androgen deprivation therapy negatively influences multiple aspects of sexual function, significantly impairing the life quality of patients diagnosed with prostate cancer. In this context, therapy through acceptance and commitment is the appropriate one, its main purpose being to change the patient’s relationship with suffering—from struggle and rejection to active acceptance and value of the present. The intervention of the psychologist or the psychotherapist is essential in decision-making counseling, using coping techniques, the clarification of personal values and the involvement of the family in the decision-making process. Oncological psychology helps the patient redefine their life goals and priorities, not just to choose a treatment. Conclusions: Sexuality and psychological health are deeply affected by prostate cancer. Psychological flexibility and emotional support can mitigate this negative impact. The integration of therapy through acceptance and commitment in the rehabilitation after treatment increases effectiveness and patient satisfaction. Full article
Show Figures

Figure 1

20 pages, 3137 KB  
Article
Mistletoe Extracts Inhibit Progressive Growth of Prostate Cancer Cells
by Sascha D. Markowitsch, Larissa Albrecht, Moritz Meiborg, Jochen Rutz, Anita Thomas, Felix K. -H. Chun, Axel Haferkamp, Eva Juengel and Roman A. Blaheta
Cells 2025, 14(19), 1535; https://doi.org/10.3390/cells14191535 - 30 Sep 2025
Abstract
Although multimodal therapeutic management has significantly improved outcome in prostate cancer (PCa) patients, treatment options for castrate-resistant disease remain challenging. Plant-derived mistletoe extracts have supported cancer patients and are, therefore, widely used as complementary medicine. However, mechanisms behind possible mistletoe benefits to PCa [...] Read more.
Although multimodal therapeutic management has significantly improved outcome in prostate cancer (PCa) patients, treatment options for castrate-resistant disease remain challenging. Plant-derived mistletoe extracts have supported cancer patients and are, therefore, widely used as complementary medicine. However, mechanisms behind possible mistletoe benefits to PCa patients remain to be explored. The present study was designed to evaluate the effect of mistletoe extracts from four different host trees (Tiliae, Populi, Salicis, and Crataegi) on the growth and proliferation of PCa cell lines in vitro. PC3, DU145, and LNCaP cells were used to evaluate tumor cell growth (MTT assay) and proliferation (BrdU incorporation assay). Clonogenicity, apoptosis, cell cycle, and cell-cycle-regulating proteins (cyclin-dependent kinases (CDKs) and cyclins) were investigated, as was CD44 standard and splice variant expression and integrin α and β receptors. SiRNA knockdown studies were employed to investigate the functional relevance of integrins. All mistletoe extracts significantly inhibited cell growth in a dose-dependent manner and cell proliferation and clonogenicity were suppressed. Populi and Salicis induced cell-cycle arrest in the G2/M phase and increased apoptosis. Both extracts down-regulated CDK1 and cyclin A and altered CD44 expression. Integrins α5 in all cell lines and α6 in DU145 and LNCaP were particularly diminished. Knocking down α5 and α6 induced cell growth inhibition in DU145. Mistletoe extracts block the growth and proliferation of PCa cells in vitro and therefore qualify for use in future animal studies to evaluate mistletoe as an adjunct to standard PCa treatment. Full article
(This article belongs to the Special Issue Natural Products and Their Derivatives Against Human Disease)
19 pages, 7006 KB  
Article
Dynamic Reprogramming of Immune-Related Signaling During Progression to Enzalutamide Resistance in Prostate Cancer
by Pengfei Xu, Huan Qu, Joy C. Yang, Fan Wei, Junwei Zhao, Menghuan Tang, Leyi Wang, Christopher Nip, Henson Li, Allen C. Gao, Kit Lam, Marc Dall'Era, Yuanpei Li and Chengfei Liu
Cancers 2025, 17(19), 3187; https://doi.org/10.3390/cancers17193187 - 30 Sep 2025
Abstract
Background: Treatment with androgen receptor (AR) signaling inhibitors, such as enzalutamide, can induce neural lineage plasticity in prostate cancer, potentially progressing to t-NEPC. However, the molecular mechanisms underlying this enzalutamide-driven plasticity, particularly the contribution of immune signaling pathways, remain poorly understood. Methods: We [...] Read more.
Background: Treatment with androgen receptor (AR) signaling inhibitors, such as enzalutamide, can induce neural lineage plasticity in prostate cancer, potentially progressing to t-NEPC. However, the molecular mechanisms underlying this enzalutamide-driven plasticity, particularly the contribution of immune signaling pathways, remain poorly understood. Methods: We analyzed transcriptomic profiles of patient samples and prostate cancer cell lines to investigate changes in immune signaling pathways. Interferon gamma (IFNγ), interferon alpha (IFNα), and interleukin 6 (IL6)-Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) signaling were assessed in enzalutamide-sensitive and -resistant prostate cancer cells. Functional assays were conducted to examine cell responsiveness to cytokine stimulation and susceptibility to STAT1 inhibition using fludarabine. Results: Immune-related pathways, including IFNγ, IFNα, IL6-JAK-STAT3, and inflammatory responses, were significantly suppressed in NEPC patient samples compared to those with castration-resistant prostate cancer (CRPC). Enzalutamide-resistant and NEPC cells exhibited markedly impaired IFNγ and IL6 signaling. In contrast, early-stage enzalutamide treatment paradoxically enhanced IFNγ and IL6 responsiveness. Transcriptomic profiling revealed coordinated upregulation of E2F target genes and activation of IFNα/IFNγ and JAK/STAT signaling pathways during early treatment. Importantly, these early-stage cells remained highly sensitive to IFNγ and IL6 stimulation and showed increased susceptibility to STAT1 inhibition by fludarabine, a sensitivity that was lost in resistant cells. Conclusions: Early enzalutamide treatment enhances immune responsiveness, while the development of resistance is associated with suppressed immune signaling and increased lineage plasticity. These results suggest a therapeutic window where combining enzalutamide with STAT inhibitors may delay or prevent lineage plasticity and resistance. Full article
Show Figures

Figure 1

30 pages, 3375 KB  
Article
Pro-Inflammatory Protein PSCA Is Upregulated in Neurological Diseases and Targets β2-Subunit-Containing nAChRs
by Mikhail A. Shulepko, Yuqi Che, Alexander S. Paramonov, Milita V. Kocharovskaya, Dmitrii S. Kulbatskii, Anisia A. Ivanova, Anton O. Chugunov, Maxim L. Bychkov, Artem V. Kirichenko, Zakhar O. Shenkarev, Mikhail P. Kirpichnikov and Ekaterina N. Lyukmanova
Biomolecules 2025, 15(10), 1381; https://doi.org/10.3390/biom15101381 - 28 Sep 2025
Abstract
Prostate stem cell antigen (PSCA) is a Ly6/uPAR protein that targets neuronal nicotinic acetylcholine receptors (nAChRs). It exists in membrane-tethered and soluble forms, with the latter upregulated in Alzheimer’s disease. We hypothesize that PSCA may be linked to a wider spectrum of neurological [...] Read more.
Prostate stem cell antigen (PSCA) is a Ly6/uPAR protein that targets neuronal nicotinic acetylcholine receptors (nAChRs). It exists in membrane-tethered and soluble forms, with the latter upregulated in Alzheimer’s disease. We hypothesize that PSCA may be linked to a wider spectrum of neurological diseases and could induce neuroinflammation. Indeed, PSCA expression is significantly upregulated in the brain of patients with multiple sclerosis, Huntington’s disease, Down syndrome, bipolar disorder, and HIV-associated dementia. To investigate PSCA’s structure, pharmacology, and inflammatory function, we produced a correctly folded water-soluble recombinant analog (ws-PSCA). In primary hippocampal neurons and astrocytes, ws-PSCA differently regulates secretion of inflammatory factors and adhesion molecules and induces pro-inflammatory responses by increasing TNFβ secretion. Heteronuclear NMR and 15N relaxation measurements reveal a classical β-structural three-finger fold with conformationally disordered loops II and III. Positive charge clustering on the molecular surface suggests the functional importance of ionic interactions by these loops. Electrophysiological studies in Xenopus oocytes point on ws-PSCA inhibition of α3β2-, high-, and low-sensitive variants of α4β2- (IC50 ~50, 27, and 15 μM, respectively) but not α4β4-nAChRs, suggesting targeting of the β2 subunit. Ensemble docking and molecular dynamics simulations predict PSCA binding to high-sensitive α4β2-nAChR at α4/β2 and β2/β2 interfaces. Complexes are stabilized by ionic and hydrogen bonds between PSCA’s loops II and III and the primary and complementary receptor subunits, including glycosyl groups. This study gives new structural and functional insights into PSCA’s interaction with molecular targets and provides clues to understand its role in the brain function and mental disorders. Full article
Show Figures

Figure 1

21 pages, 1015 KB  
Review
Research Advances in Cancer-Associated Fibroblasts in Prostate Cancer Progression
by Zhonghao Tang, Si Shen, Chenwei Gu, Sixin Li, Yan Qin and Yuanyuan Mi
Biomolecules 2025, 15(10), 1369; https://doi.org/10.3390/biom15101369 - 26 Sep 2025
Abstract
The tumor microenvironment (TME) is crucial for tumor growth and progression, within which cancer-associated fibroblasts (CAFs) play a central role in regulating cancer cell proliferation, metastasis, and therapy resistance through various mechanisms. Although early-stage prostate cancer (PCa) has a high cure rate, advanced [...] Read more.
The tumor microenvironment (TME) is crucial for tumor growth and progression, within which cancer-associated fibroblasts (CAFs) play a central role in regulating cancer cell proliferation, metastasis, and therapy resistance through various mechanisms. Although early-stage prostate cancer (PCa) has a high cure rate, advanced disease often becomes difficult to manage due to resistance to standard therapies such as androgen deprivation therapy (ADT). Therefore, a deep understanding of the interaction mechanisms between CAFs and PCa cells is essential for developing novel therapeutic strategies targeting resistant advanced PCa. This review systematically summarizes key signaling pathways and molecular mechanisms through which CAFs promote PCa progression, as recently discovered, evaluates the potential of CAFs as prognostic biomarkers, and discusses novel CAF-based therapeutic targets and intervention strategies for PCa. Full article
(This article belongs to the Special Issue Molecular Advances in Drug Resistance and Novel Therapies for Cancer)
Show Figures

Figure 1

11 pages, 1071 KB  
Article
Homotypic Targeting of [89Zr]Zr-Oxine Labeled PC3 and 4T1 Cells in Tumor-Bearing Mice
by Volkan Tekin, Noel E. Archer, Solana R. Fernandez, Hailey A. Houson, Jennifer L. Bartels and Suzanne E. Lapi
Pharmaceutics 2025, 17(10), 1259; https://doi.org/10.3390/pharmaceutics17101259 - 26 Sep 2025
Abstract
Background/Objectives: Homotypic targeting refers to the ability of cells to preferentially interact with other cells of the same type. An understanding of how cells use homotypic targeting (self-homing) characteristics for tumor-targeting purposes may aid in the effective delivery of radionuclides or other [...] Read more.
Background/Objectives: Homotypic targeting refers to the ability of cells to preferentially interact with other cells of the same type. An understanding of how cells use homotypic targeting (self-homing) characteristics for tumor-targeting purposes may aid in the effective delivery of radionuclides or other drugs for imaging or therapeutic applications. Additionally, studies investigating the targeting properties of cells from the same lineage may shed light on this interesting mechanism, allowing it to be harnessed for other applications. The objective of this study was to assess the tumor-self targeting potential of PC3 prostate cancer and 4T1 breast cancer cells using a direct cell labeling technique, with a focus on evaluation of cellular labeling efficiency, cell viability, cellular efflux, and in vivo tumor-self targeting capability using both identical and dissimilar tumor models. Methods: [89Zr]Zr-oxine was prepared and utilized for the labeling of PC3 and 4T1 cells. Following the assessment of cell labeling efficacy, viability, and efflux, PET/CT imaging and biodistribution studies were conducted with [89Zr]Zr-oxine labeled PC3 and 4T1 cells in PC3 and 4T1 tumor-bearing mice models. Results: Both PC3 cells and 4T1 cells were radiolabeled with [89Zr]oxine, with PC3 cells illustrating a higher labeling efficiency (86.55 ± 0.38%) than 4T1 cells (46.95 ± 1.47%). Notably, radiolabeled PC3 cells illustrated significant uptake in PC3 tumors (7.54 ± 1.07%ID/gram at 24 h and 6.95 ± 3.56%ID/gram at 48 h) with lower tumor uptake in the 4T1 xenograft model (1.79 ± 0.29%ID/gram at 24 h and 1.42 ± 0.71%ID/gram at 48 h), illustrating the potential of self-targeting. Conclusions: Both PC3 and 4T1 cells followed a similar pattern of biodistribution, with labeled PC3 cells demonstrating lower blood retention and reduced uptake in non-target organs such as lungs and heart. Taken together, these results may indicate that PC3 cells illustrate homotypic targeting, warranting further investigation of this phenomenon. Full article
(This article belongs to the Special Issue Cell-Mediated Delivery Systems)
Show Figures

Graphical abstract

15 pages, 3484 KB  
Article
Stefin A Regulation of Cathepsin B Expression and Localization in Cancerous and Non-Cancerous Cells
by Anastasiia O. Syrocheva, Ekaterina P. Kolesova, Alessandro Parodi and Andrey A. Zamyatnin
Int. J. Mol. Sci. 2025, 26(19), 9321; https://doi.org/10.3390/ijms26199321 - 24 Sep 2025
Viewed by 52
Abstract
Cathepsin B (CTSB), a lysosomal cysteine protease, plays pivotal roles in cellular homeostasis and pathology, including cancer progression. This study investigates the regulatory interplay between CTSB and Stefin A (STFA), an endogenous inhibitor of cysteine proteases, in renal and prostate cancer cells. Using [...] Read more.
Cathepsin B (CTSB), a lysosomal cysteine protease, plays pivotal roles in cellular homeostasis and pathology, including cancer progression. This study investigates the regulatory interplay between CTSB and Stefin A (STFA), an endogenous inhibitor of cysteine proteases, in renal and prostate cancer cells. Using plasmid-based overexpression and silencing systems, we demonstrated that overexpressing STFA significantly reduces CTSB activity and protein levels, while silencing STFA leads to elevated CTSB activity and expression in cancer cells but not in non-cancerous cells (embryonic kidney cells—Hek293T and endothelial cells—EA.hy926). Furthermore, STFA modulates the subcellular distribution of CTSB, with STFA overexpression reducing nuclear CTSB levels and silencing inducing cytoplasmic accumulation in cancer cells. Colocalization analysis confirms a direct interaction between STFA and CTSB, highlighting the spatial coordination necessary for effective protease inhibition. These findings underscore the critical role of the CTSB-STFA axis in maintaining proteolytic balance and suggest potential therapeutic strategies targeting this interaction in renal carcinoma and other cancers. Full article
(This article belongs to the Special Issue Molecular Mechanisms and New Markers of Cancer)
Show Figures

Figure 1

12 pages, 1242 KB  
Brief Report
Functional HER1/HER2-Expressing Murine Tumor Models for Preclinical Evaluation of Targeted Therapies
by Talia Fundora-Barrios, Amanda R. Hechavarría-Bajuelo, Lisset Chao García, Miguel Angel Gonzalez-Cruz, Najara Gonzalez-Suarez, Gretchen Bergado-Baez and Belinda Sánchez-Ramírez
Receptors 2025, 4(4), 18; https://doi.org/10.3390/receptors4040018 - 24 Sep 2025
Viewed by 69
Abstract
Background: HER1 and HER2 are critical receptors involved in tumorigenesis and the development of targeted therapies for various carcinomas. However, most antibodies and drugs currently in development do not recognize murine orthologs, which restricts their evaluation in immunocompetent models. Methods: We generated nine [...] Read more.
Background: HER1 and HER2 are critical receptors involved in tumorigenesis and the development of targeted therapies for various carcinomas. However, most antibodies and drugs currently in development do not recognize murine orthologs, which restricts their evaluation in immunocompetent models. Methods: We generated nine tumor models through the lentiviral transduction of murine prostate (RM1), lung (3LL-D122), and breast (4T1) carcinoma cell lines, subsequently validating them in immunocompetent BALB/c and C57BL/6 hosts. Receptor expression and functionality were characterized using flow cytometry, immunoblotting, proliferation assays, and therapeutic sensitivity testing. Results: Transduced cells exhibited stable membrane expression of HER1/HER2 and ligand-induced phosphorylation, confirming receptor functionality. In all three tumor models generated, the expression of HER1 and/or HER2 significantly enhanced cell proliferation compared to parental lines. Furthermore, treatment with specific monoclonal antibodies and the tyrosine kinase inhibitor markedly reduced the viability of cells expressing HER1 and/or HER2, without affecting negative controls. Conclusions: These models provide a robust and reproducible platform for the preclinical evaluation of HER1/HER2-targeted therapies in immunocompetent hosts. Although the current model relies on subcutaneous implantation and does not fully replicate the native tumor microenvironment, it represents a crucial first step toward the development of orthotopic and immunologically relevant models for translational cancer research. Full article
(This article belongs to the Collection Receptors: Exceptional Scientists and Their Expert Opinions)
Show Figures

Graphical abstract

12 pages, 830 KB  
Article
Can PSMA-Targeting Radiopharmaceuticals Be Useful for Detecting Brain Metastasis of Various Tumors Using Positron Emission Tomography?
by Esra Arslan, Nurhan Ergül, Rahime Şahin, Ediz Beyhan, Özge Erol Fenercioğlu, Yeşim Karagöz, Arzu Algün Gedik, Yakup Bozkaya and Tevfik Fikret Çermik
Cancers 2025, 17(18), 3088; https://doi.org/10.3390/cancers17183088 - 22 Sep 2025
Viewed by 169
Abstract
Objective: The high expression of prostate-specific membrane antigen (PSMA) associated with neovascularization in non-prostatic malignant tumors and metastatic lesions has been documented in many studies. By taking advantage of the absence of PSMA-related background activity in brain tissue, in recent years, PSMA has [...] Read more.
Objective: The high expression of prostate-specific membrane antigen (PSMA) associated with neovascularization in non-prostatic malignant tumors and metastatic lesions has been documented in many studies. By taking advantage of the absence of PSMA-related background activity in brain tissue, in recent years, PSMA has been used for the imaging of glial tumors, especially for postoperative follow-up. The aim of this prospective study was to investigate the diagnostic value of 68Ga-PSMA-11 PET/CT by comparing 68Ga-PSMA-11 PET/CT, 18F-FDG PET/CT, and MRI findings in patients with brain metastases (BM). Materials and Method: In this prospective study, 27 cases, 11 female and 16 male, with a mean age of 59.48 ± 12.21 years, were included. Patients diagnosed with BM on 18F-FDG PET/CT or CT/MRI at initial diagnosis or in the follow-up period were included in the study. PET findings of BM lesions obtained from 18F-FDG and 68Ga-PSMA-11 PET/CT imaging, demographic characteristics, histopathological data of the primary foci, and other clinical features were evaluated together. Results: Twenty-four (89%) patients were included in the study for restaging, two (7%) patients for local recurrence assessment, and one (4%) patient for local recurrence and suspicion of additional lesions. The indications for 18F-FDG PET/CT were breast carcinoma for 37% (n:10), followed by lung carcinoma for 26% (n:7), colorectal adenocarcinoma for 14% (n:4), squamous cell larynx carcinoma for 7% (n:2), gastric signet ring cell carcinoma for 4% (n:1), pancreatic NET3 for 4% (n:1), thyroid papillary carcinoma for 4% (n:1), and malignant melanoma for 4% (n:1). In total, 26/27 included patients had PSMA-positive brain metastases but only one patient had PSMA-negative brain metastases with 68Ga-PSMA-11 PET/CT imaging. This patient was followed with a diagnosis of primary larynx squamous carcinoma and had a mass suspected of brain metastases. Further tests and an MRI revealed that the lesion in this patient was a hemorrhagic metastasis. The smallest metastatic focus on 68Ga-PSMA-11 PET/CT imaging was 0.22 cm, also confirmed by MRI (range: 0.22–2.81 cm). The mean ± SD SUVmax of the BM lesions was 17.9 ± 8.6 and 6.8 ± 5.2 on 18F-FDG PET/CT and 68Ga-PSMA-11 PET/CT imaging, respectively. Metastatic foci that could not be detected by 18F-FDG PET/CT imaging were successfully detected with 68Ga-PSMA-11 PET/CT imaging in 11 cases (42%). The distribution and number of metastatic lesions observed on cranial MRI and 68Ga-PSMA-11 PET/CT were compatible with each other for all patients. Immunohistochemical staining was performed in the primary tumor of 10 (38%) cases, and positive IHC staining with PSMA was detected in 5 patients. In addition, positive IHC staining with PSMA was detected in all of the four surgically excised brain metastatic tumor foci. Conclusions: In this study,68Ga-PSMA-11 PET/CT appears to be superior to 18F-FDG in detecting BM from various tumors, largely due to its high expression associated with neovascularization and the absence of PSMA expression in normal brain parenchyma. This lack of physiological uptake in healthy brain tissue provides excellent tumor-to-background contrast, further supporting the advantage of 68Ga-PSMA-11 over 18F-FDG for BM imaging. However, larger studies are required to confirm these findings, particularly through comparisons across tumor types and histopathological subgroups, integrating PSMA immunohistochemistry (IHC) scores with 68Ga-PSMA-11 uptake levels. Beyond its diagnostic potential, our results may also inform PSMA-targeted therapeutic strategies, offering new perspectives for the management of patients with brain metastases from diverse primary tumors. Full article
Show Figures

Figure 1

14 pages, 1621 KB  
Article
Cytotoxicity of Esculetin Compared with Vinblastine and Paclitaxel in PC-3 Prostate Cancer Cells
by Ana I. García-Pérez, Virginia Rubio, Angel Herráez, Lilian Puebla and José C. Diez
Curr. Oncol. 2025, 32(9), 526; https://doi.org/10.3390/curroncol32090526 - 20 Sep 2025
Viewed by 150
Abstract
Background/Objectives: Metastatic prostate cancer is among the therapy-resistant human neoplasms. PC-3 is a commonly used experimental cell line that does not express androgen receptors. We compared the cytotoxicity of esculetin with that of vinblastine and paclitaxel on prostatic tumour PC-3 cells. Methods: Cells [...] Read more.
Background/Objectives: Metastatic prostate cancer is among the therapy-resistant human neoplasms. PC-3 is a commonly used experimental cell line that does not express androgen receptors. We compared the cytotoxicity of esculetin with that of vinblastine and paclitaxel on prostatic tumour PC-3 cells. Methods: Cells were treated with either esculetin (100 or 250 μM), vinblastine (50 μM) or paclitaxel (100 or 200 μM) for 19 to 72 h. Cells were assessed for metabolic viability, membrane integrity, DNA fragmentation and cell cycle analysis. Apoptosis was checked with annexin and propidium iodide. Results: Esculetin decreased the metabolic activity of PC-3 cells in a time- and concentration-dependent way. The metabolic activity of vinblastine- and paclitaxel-treated cells did not show time-dependence. Cells treated with 250 µM esculetin for 48 or 72 h showed apoptosis levels similar to those produced by 50 µM vinblastine at these incubation times or by 200 µM paclitaxel at 19 h. Vinblastine and paclitaxel produced cell cycle arrest in the G2/M phase after incubation for 19 h. In contrast, esculetin did not significantly affect the cell cycle. Conclusions: A differential action of esculetin on PC-3 prostate cells may be inferred. This may be relevant for novel therapies against resistant prostate cancer. Full article
(This article belongs to the Collection New Insights into Prostate Cancer Diagnosis and Treatment)
Show Figures

Graphical abstract

24 pages, 2844 KB  
Article
Theranostic Potential of a New 64Cu-Labeled NOTA-R954 Peptide Conjugate for Kinin B1R Expressing Prostate Cancer
by Sadaf Ghanaatgar Kasbi, Martin Savard, Frédéric Couture, Céléna Dubuc, Véronique Dumulon-Perreault, Marie-Edith Nepveu-Traversy, Samia Ait-Mohand, Robert Sabbagh, Sameh Geha, Brigitte Guérin, Yves Dory and Fernand Gobeil
Pharmaceutics 2025, 17(9), 1215; https://doi.org/10.3390/pharmaceutics17091215 - 18 Sep 2025
Cited by 1 | Viewed by 401
Abstract
Background/Objectives: This study explores the potential of the inducible G protein-coupled kinin B1 receptor (B1R) as a target for the diagnosis and treatment of prostate cancer (PCa) and aims to develop the first theranostic agent targeting hB1R for both molecular imaging and [...] Read more.
Background/Objectives: This study explores the potential of the inducible G protein-coupled kinin B1 receptor (B1R) as a target for the diagnosis and treatment of prostate cancer (PCa) and aims to develop the first theranostic agent targeting hB1R for both molecular imaging and radionuclide therapy. Methods: B1R expression was analyzed via qPCR and immunohistochemistry in human PCa cells and tissues specimens. A novel 64Cu/NOTA-conjugated peptide analog of the potent B1R antagonist R954 was synthetized and evaluated in vitro and in vivo. Results: B1R was confirmed to be expressed (RNA, protein) by varying degrees in all PCa cell lines and tissues investigated, with protein level significantly correlating with tumor grades. This finding was supported by similar analyses from the TCGA and MSKCC databases. In vitro, the 64Cu/NOTA-βAla-R954 conjugate showed nanomolar affinity/potency at hB1R, complete plasma stability over 24 h, significant cellular uptake (up to 33% of ID at 24 h), and dose-dependent anti-clonal growth effects. In vivo, the radioconjugate remained stable in circulation for up to 90 min and was primarily excreted intact via the kidneys following IV administration. Intravenous 64Cu/NOTA-βAla-R954 (7.5 MBq) effectively detected subcutaneous PCa xenografts via µPET imaging in male athymic nude mice. At a single higher dose (65 MBq; 50 µg/kg), it significantly reduced tumor growth without observable toxicity. This antitumor effect was associated with increased apoptosis (active caspase-3) and reduced proliferation (Ki67), as shown by immunohistochemistry. In contrast, the nonradioactive NatCu/NOTA-βAla-R954 had no therapeutic effect at the same dose. Conclusions: Our findings provide proof-of-concept for the potential theranostic use of 64Cu/NOTA-R954 in PCa, and potentially other types of B1R-positive solid cancers. Full article
Show Figures

Figure 1

Back to TopTop