Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (133)

Search Parameters:
Keywords = neuroendocrine prostate cancer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
32 pages, 1319 KiB  
Review
Effects of Targeted Radionuclide Therapy on Cancer Cells Beyond the Ablative Radiation Dose
by Guillermina Ferro-Flores, Erika Azorín-Vega, Blanca Ocampo-García, Myrna Luna-Gutiérrez, Pedro Cruz-Nova and Laura Meléndez-Alafort
Int. J. Mol. Sci. 2025, 26(14), 6968; https://doi.org/10.3390/ijms26146968 - 20 Jul 2025
Viewed by 657
Abstract
Targeted radionuclide therapy (TRT) utilizes radiopharmaceuticals to deliver radiation directly to cancer cells while sparing healthy tissues. Beyond the absorbed dose of ablative radiation, TRT induces non-targeted effects (NTEs) that significantly enhance its therapeutic efficacy. These effects include radiation-induced bystander effects (RIBEs), abscopal [...] Read more.
Targeted radionuclide therapy (TRT) utilizes radiopharmaceuticals to deliver radiation directly to cancer cells while sparing healthy tissues. Beyond the absorbed dose of ablative radiation, TRT induces non-targeted effects (NTEs) that significantly enhance its therapeutic efficacy. These effects include radiation-induced bystander effects (RIBEs), abscopal effects (AEs), radiation-induced genomic instability (RIGI), and adaptive responses, which collectively influence the behavior of cancer cells and the tumor microenvironment (TME). TRT also modulates immune responses, promoting immune-mediated cell death and enhancing the efficacy of combination therapies, such as the use of immune checkpoint inhibitors. The molecular mechanisms underlying TRT involve DNA damage, oxidative stress, and apoptosis, with repair pathways like homologous recombination (HR) and non-homologous end joining (NHEJ) playing critical roles. However, challenges such as tumor heterogeneity, hypoxia, and radioresistance limit the effectiveness of this approach. Advances in theranostics, which integrate diagnostic imaging with TRT, have enabled personalized treatment approaches, while artificial intelligence and improved dosimetry offer potential for treatment optimization. Despite the significant survival benefits of TRT in prostate cancer and neuroendocrine tumors, 30–40% of patients remain unresponsive, which highlights the need for further research into molecular pathways, long-term effects, and combined therapies. This review outlines the dual mechanisms of TRT, direct toxicity and NTEs, and discusses strategies to enhance its efficacy and expand its use in oncology. Full article
(This article belongs to the Special Issue Targeted Therapy of Cancer: Innovative Drugs and Molecular Tools)
Show Figures

Figure 1

20 pages, 12298 KiB  
Article
Impact of Metastatic Microenvironment on Physiology and Metabolism of Small Cell Neuroendocrine Prostate Cancer Patient-Derived Xenografts
by Shubhangi Agarwal, Deepti Upadhyay, Jinny Sun, Emilie Decavel-Bueff, Robert A. Bok, Romelyn Delos Santos, Said Al Muzhahimi, Rosalie Nolley, Jason Crane, John Kurhanewicz, Donna M. Peehl and Renuka Sriram
Cancers 2025, 17(14), 2385; https://doi.org/10.3390/cancers17142385 - 18 Jul 2025
Viewed by 442
Abstract
Background: Potent androgen receptor pathway inhibitors induce small cell neuroendocrine prostate cancer (SCNC), a highly aggressive subtype of metastatic androgen deprivation-resistant prostate cancer (ARPC) with limited treatment options and poor survival rates. Patients with metastases in the liver have a poor prognosis relative [...] Read more.
Background: Potent androgen receptor pathway inhibitors induce small cell neuroendocrine prostate cancer (SCNC), a highly aggressive subtype of metastatic androgen deprivation-resistant prostate cancer (ARPC) with limited treatment options and poor survival rates. Patients with metastases in the liver have a poor prognosis relative to those with bone metastases alone. The mechanisms that underlie the different behavior of ARPC in bone vs. liver may involve factors intrinsic to the tumor cell, tumor microenvironment, and/or systemic factors, and identifying these factors is critical to improved diagnosis and treatment of SCNC. Metabolic reprogramming is a fundamental strategy of tumor cells to colonize and proliferate in microenvironments distinct from the primary site. Understanding the metabolic plasticity of cancer cells may reveal novel approaches to imaging and treating metastases more effectively. Methods: Using magnetic resonance (MR) imaging and spectroscopy, we interrogated the physiological and metabolic characteristics of SCNC patient-derived xenografts (PDXs) propagated in the bone and liver, and used correlative biochemical, immunohistochemical, and transcriptomic measures to understand the biological underpinnings of the observed imaging metrics. Results: We found that the influence of the microenvironment on physiologic measures using MRI was variable among PDXs. However, the MR measure of glycolytic capacity in the liver using hyperpolarized 13C pyruvic acid recapitulated the enzyme activity (lactate dehydrogenase), cofactor (nicotinamide adenine dinucleotide), and stable isotope measures of fractional enrichment of lactate. While in the bone, the congruence of the glycolytic components was lost and potentially weighted by the interaction of cancer cells with osteoclasts/osteoblasts. Conclusion: While there was little impact of microenvironmental factors on metabolism, the physiological measures (cellularity and perfusion) are highly variable and necessitate the use of combined hyperpolarized 13C MRI and multiparametric (anatomic, diffusion-, and perfusion- weighted) 1H MRI to better characterize pre-treatment tumor characteristics, which will be crucial to evaluate treatment response. Full article
(This article belongs to the Special Issue Magnetic Resonance in Cancer Research)
Show Figures

Figure 1

27 pages, 4850 KiB  
Review
Unlocking the Role of OCT4 in Cancer Lineage Plasticity: A Cross-Cancer Perspective with an Emphasis on Prostate Cancer
by Mohammad Esfini Farahani, Yanquan Zhang, Amos Olalekan Akinyemi, Fatemeh Seilani, Md Rakibul Alam and Xiaoqi Liu
Biomedicines 2025, 13(7), 1642; https://doi.org/10.3390/biomedicines13071642 - 4 Jul 2025
Viewed by 631
Abstract
Prostate cancer (PCa) is a highly heterogeneous disease, with castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) representing its most aggressive and therapy-resistant forms. Emerging evidence indicates that lineage plasticity—driven by key transcription factors such as Octamer Binding Factor 4 (OCT4)—plays a [...] Read more.
Prostate cancer (PCa) is a highly heterogeneous disease, with castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) representing its most aggressive and therapy-resistant forms. Emerging evidence indicates that lineage plasticity—driven by key transcription factors such as Octamer Binding Factor 4 (OCT4)—plays a crucial role in therapeutic resistance and disease progression. OCT4, in coordination with SOX2 and NANOG, acts as a master regulator of stemness and is frequently upregulated in prostate cancer stem cells (PCSCs). This upregulation contributes to tumor initiation, metastasis, and resistance to both androgen deprivation therapy (ADT) and chemotherapy. In this review, we explore the role of OCT4 in mediating lineage plasticity in prostate cancer, with particular emphasis on its involvement in treatment resistance and neuroendocrine differentiation. We also examine therapeutic strategies aimed at targeting OCT4 directly, such as microRNA-mediated suppression, small-molecule inhibitors, and suicide gene therapy, as well as indirect approaches that modulate OCT4 expression via FGFR and NF-κB signaling pathways. While these strategies offer promising avenues, challenges such as adaptive resistance and the intricate signaling networks within PCSCs remain significant hurdles. A deeper understanding of the molecular mechanisms underlying OCT4-driven plasticity may pave the way for novel therapeutic approaches and improved outcomes in advanced prostate cancer. Full article
(This article belongs to the Special Issue Molecular Biomarkers of Tumors: Advancing Genetic Studies)
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 827
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

25 pages, 2811 KiB  
Review
68Ga Extemporaneous Preparations in Radiopharmacy
by Marzia Rizzello, Anna Pacelli, Maria Domenica De Bari, Annalisa Cutrignelli, Rosa Maria Iacobazzi, Antonio Lopalco and Nunzio Denora
Pharmaceutics 2025, 17(7), 802; https://doi.org/10.3390/pharmaceutics17070802 - 20 Jun 2025
Viewed by 691
Abstract
Gallium-68 (68Ga) radiopharmaceuticals are increasingly used in nuclear medicine due to their rapid production capabilities and exceptional specificity in molecular imaging applications. Several of these tracers have demonstrated remarkable clinical efficacy across various oncological conditions, including prostate cancer, neuro-endocrine tumours, and [...] Read more.
Gallium-68 (68Ga) radiopharmaceuticals are increasingly used in nuclear medicine due to their rapid production capabilities and exceptional specificity in molecular imaging applications. Several of these tracers have demonstrated remarkable clinical efficacy across various oncological conditions, including prostate cancer, neuro-endocrine tumours, and cancers expressing fibroblast activation protein. Commercial kits allow the use of the standardised production protocol, but extemporaneous preparations are the economic and flexible alternatives, particularly within hospital-based radiopharmacy settings. However, such preparations need meticulous conformity to quality control measures and regulation to ensure safety and effectiveness. This review provides an analysis of current methodologies employed in 68Ga extemporaneous preparations and examines pertinent regulatory frameworks. Further clinical validation trials and technical advancement remain essential to facilitate the routine clinical practice’s widespread usage and long-term feasibility of such preparations. Full article
Show Figures

Graphical abstract

20 pages, 3005 KiB  
Article
Phenotypic Plasticity and Androgen Receptor Bypass Drive Cross-Resistance to Apalutamide in Castration-Resistant Prostate Cancer Cell Models
by Iris Simon, Jose Manuel Sanchez-Manas, Sonia Perales, Gonzalo Martinez-Navajas, Jorge Ceron-Hernandez and Pedro J. Real
Int. J. Mol. Sci. 2025, 26(13), 5939; https://doi.org/10.3390/ijms26135939 - 20 Jun 2025
Viewed by 540
Abstract
The treatment of choice for prostate cancer is androgen deprivation (ADT) and novel hormonal agents such as Abiraterone, Enzalutamide, or Apalutamide. Initially, this therapy is highly effective, but a significant challenge arises as most patients eventually develop resistance, resulting in castration-resistant prostate cancer [...] Read more.
The treatment of choice for prostate cancer is androgen deprivation (ADT) and novel hormonal agents such as Abiraterone, Enzalutamide, or Apalutamide. Initially, this therapy is highly effective, but a significant challenge arises as most patients eventually develop resistance, resulting in castration-resistant prostate cancer (CRPC). Furthermore, the sequential use of these drugs can lead to cross-resistance, diminishing their efficacy. Tumor heterogeneity plays a pivotal role in the development of resistance to different treatments. This study utilized cellular models of CRPC to assess the response to Apalutamide when it was administered as a second- or third-line treatment. Functional and genetic analyses were conducted in various CRPC cell models exposed to Apalutamide. These analyses included real-time cell monitoring assays, flow cytometry, clonogenicity assays, and RT-qPCR. CRPC cell models were capable of continued proliferation, maintained cell cycle profiles similar to those of untreated cells, and retained their clonogenic potential. Cross-resistance to Apalutamide in models of ADT, ADT plus Enzalutamide, or Abiraterone resistance did not correlate with the expression levels of AR-V7 and AR-V9 variants. Gene expression analysis of resistant prostate cancer cell lines revealed that treatment with Apalutamide induced the emergence of more aggressive phenotypes, including cancer stem cells or neuroendocrine differentiation profiles. Most CRPC cell models developed cross-resistance to Apalutamide and were able to proliferate and retain their clonogenic capability. Apalutamide resistance was not linked to the expression of AR-V7 or AR-V9 variants but was instead associated to bypass of AR signaling pathway and the emergence of more aggressive expression profiles. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

34 pages, 1078 KiB  
Review
Radionuclide-Labeled Biomaterials: A Novel Strategy for Tumor-Targeted Therapy
by Shu Zhang, Aiyue Zhang, Xunhao Qi, Zongtai Han, Luqi Song, Jiayu Zhou, Guanglin Wang, Ran Zhu and Jianguo Li
Biomimetics 2025, 10(6), 394; https://doi.org/10.3390/biomimetics10060394 - 11 Jun 2025
Viewed by 1159
Abstract
This paper presents a comprehensive review of recent advancements in radionuclide-labeled biomaterials for cancer therapy, with a particular focus on the characteristics, production methods, and labeling techniques of α-particle, β-particle, and Auger electron-based radiotherapy. It explores innovative strategies for targeted delivery systems and [...] Read more.
This paper presents a comprehensive review of recent advancements in radionuclide-labeled biomaterials for cancer therapy, with a particular focus on the characteristics, production methods, and labeling techniques of α-particle, β-particle, and Auger electron-based radiotherapy. It explores innovative strategies for targeted delivery systems and highlights the advantages of theranostics and combination therapies. The application of radionuclide-labeled biomaterials in various cancer types, including prostate cancer, breast cancer, neuroendocrine tumors, gliomas, and melanoma, is systematically summarized. Furthermore, the article critically examines current technological bottlenecks and challenges in clinical translation, while proposing future directions such as AI-assisted dose optimization and multimodal combination therapies. This review provides essential theoretical foundations and practical insights to facilitate the clinical translation of radionuclide-labeled biomaterials. Full article
(This article belongs to the Special Issue Biological Materials and Bioinspired Designs)
Show Figures

Graphical abstract

9 pages, 786 KiB  
Communication
Local Therapy Can Enhance the Prognosis of Certain Patients with Pathologically Diagnosed Neuroendocrine Prostate Carcinoma
by Shoichi Kimura, Naoki Terada, Shinnya Soumiya, Takayuki Goto, Hiromitsu Negoro, Shoichiro Mukai, Osamu Ogawa, Shusuke Akamatsu, Takashi Kobayashi, Atsuro Sawada and Toshiyuki Kamoto
Life 2025, 15(5), 797; https://doi.org/10.3390/life15050797 - 17 May 2025
Cited by 1 | Viewed by 594
Abstract
Neuroendocrine prostate cancer (NEPC) has a poor prognosis. We performed a retrospective analysis of the factors contributing to survival in patients with histologically diagnosed NEPC. Patients pathologically diagnosed with NEPC between 2007 and 2018 were retrospectively analyzed. Overall survival (OS) from the time [...] Read more.
Neuroendocrine prostate cancer (NEPC) has a poor prognosis. We performed a retrospective analysis of the factors contributing to survival in patients with histologically diagnosed NEPC. Patients pathologically diagnosed with NEPC between 2007 and 2018 were retrospectively analyzed. Overall survival (OS) from the time of the initial prostate cancer diagnosis was evaluated using the Kaplan–Meier method. Cox proportional hazards analyses were performed to evaluate the association of OS with variables including the presence of metastasis, receipt of local therapy, and disease classification (primary NEPC [p-NEPC] or treatment-related NEPC [t-NEPC]). Among 32 patients (p-NEPC, 22; t-NEPC, 10), distant metastases were identified in 25 (78%) patients, and local therapies including radical prostatectomy and local radiotherapy were provided to 21 (66%) patients. In the univariate Cox proportional hazard analyses, patients who received local therapy had a significantly lower risk of death than those who did not receive local therapy (hazard ratio = 0.284, 95% confidence interval = 0.109–0.738, p = 0.01). OS was significantly longer for patients receiving local therapy than for those who did not receive local therapy (36 months vs. 13 months, p = 0.0058). Our findings suggest the potential benefit of local therapy in the treatment of NEPC. Full article
(This article belongs to the Special Issue Prostate Cancer: 4th Edition)
Show Figures

Figure 1

15 pages, 1803 KiB  
Article
Pilot Investigation on Markers of Bone Metabolism, Angiogenesis, and Neuroendocrine Activity as Potential Predictors of Survival of Metastatic Prostate Cancer Patients with Bone Metastases
by Maria Angels Ortiz, Georgia Anguera, Elisabet Cantó, Jose Alejandre, Josefina Mora, Ruben Osuna-Gómez, Maria Mulet, Pradip Mora, Assumpta Antonijuan, Sofia Sánchez, Ona Ramírez, Vanessa Orantes, Pablo Maroto and Silvia Vidal
Int. J. Mol. Sci. 2025, 26(10), 4669; https://doi.org/10.3390/ijms26104669 - 13 May 2025
Viewed by 713
Abstract
Prostate cancer with bone metastasis exhibits significant heterogeneity, complicating prognosis, and treatment. This study explores the potential of plasma, serum, and urine biomarkers to stratify patients and evaluate their prognostic value. Using two-step clustering, we analyzed baseline levels of Platelet-derived growth factor-BB (PDGF-BB), [...] Read more.
Prostate cancer with bone metastasis exhibits significant heterogeneity, complicating prognosis, and treatment. This study explores the potential of plasma, serum, and urine biomarkers to stratify patients and evaluate their prognostic value. Using two-step clustering, we analyzed baseline levels of Platelet-derived growth factor-BB (PDGF-BB), Insulin-like growth factor-binding protein 1 (IGFBP-1), Bone Morphogenetic Protein 2 (BMP-2), Vascular endothelial growth factor (VEGF) (plasma and urine), prostate-specific antigen (PSA), neuron-specific enolase (NSE), chromogranin A (CgA) and bone-specific alkaline phosphatase (BAP) (serum) and creatinine (Cr), and type I collagen-cross-linked N telopeptide (NTx) (urine) in 29 patients with prostate cancer and bone metastasis. Longitudinal biomarker dynamics were assessed at baseline, 6 months, and 12 months. Clinical outcomes were evaluated using Kaplan–Meier and multivariate analyses. Three distinct groups (C1, C2, and C3) were identified. C1 exhibited elevated pPDGF-BB and pVEGF levels, C3 had increased pBAP and uNTx/Cr, and C2 showed lower biomarker levels. Prior treatments influenced biomarker levels, with bisphosphonates reducing bone turnover markers and radiotherapy correlating with long-term changes in growth factors. Longitudinal analysis revealed unique biomarker dynamics within each group, with a tendency for pPDGF-BB and pVEGF levels to decrease over time in C1, and distinct trends in uNTx/Cr between groups. Despite individual biomarkers failing to predict survival, C3 patients demonstrated significantly worse survival than C1 and C2. Molecular clustering of peripheral blood and urinary biomarkers identifies distinct subgroups with metastatic castration-resistant prostate cancer patients outperforming traditional models in outcome prediction and supporting its potential for personalized treatment and prognosis. Full article
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 1671
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

27 pages, 19579 KiB  
Article
Atypical Pelvic Tumors in Children
by Paulina Sobieraj and Monika Bekiesińska-Figatowska
Cancers 2025, 17(4), 619; https://doi.org/10.3390/cancers17040619 - 12 Feb 2025
Viewed by 1811
Abstract
Due to the complex anatomy of the pelvis, various tumors may arise in this region. Some of these tumors are well known and have distinctive features that allow them to be identified by magnetic resonance imaging (MRI). These include sacrococcygeal teratoma (SCT), the [...] Read more.
Due to the complex anatomy of the pelvis, various tumors may arise in this region. Some of these tumors are well known and have distinctive features that allow them to be identified by magnetic resonance imaging (MRI). These include sacrococcygeal teratoma (SCT), the most prevalent congenital tumor in children, often diagnosed prenatally and most frequently occurring in this anatomical location, and ovarian teratoma, which in its mature form is the most common ovarian neoplasm in children and adolescents. Additionally, rhabdomyosarcoma (RMS), commonly found in the bladder in both genders and in the prostate in males, and Ewing sarcoma (ES), affecting the flat bones of the pelvis, are relatively common tumors. In this study, selected atypical pelvic tumors in children are presented. Most of them are tumors of the reproductive system, such as cervical cancer, small cell neuroendocrine carcinoma of the ovary, ES/primitive neuroectodermal tumor (PNET) of the ovary, diffuse large B-cell lymphoma (DLBCL) of the ovaries and ovarian Sertoli–Leydig cell tumor (SLCT) with RMS due to DICER1 syndrome. Additionally, tumors originating from the nervous system, including neuroblastoma (NBL) and plexiform neurofibroma (pNF), associated and not associated with neurofibromatosis type 1 (NF1), are discussed. Furthermore, Rosai–Dorfman disease involving the pelvic and inguinal lymph nodes is presented. By reviewing the literature and presenting our cases, we tried to find radiological features of individual tumors that would bring the radiologist closer to the correct diagnosis, ensuring the implementation of appropriate treatment. However, the MR images cannot be considered in isolation. Additional patient data, such as the clinical picture, comorbidities/syndromes, and laboratory test results, are necessary. Full article
(This article belongs to the Section Pediatric Oncology)
Show Figures

Figure 1

28 pages, 5502 KiB  
Article
p27Kip1 and Tumors: Characterization of CDKN1B Variants Identified in MEN4 and Breast Cancer
by Debora Bencivenga, Emanuela Stampone, Jahanzaib Azhar, Daniela Parente, Waqar Ali, Vitale Del Vecchio, Fulvio Della Ragione and Adriana Borriello
Cells 2025, 14(3), 188; https://doi.org/10.3390/cells14030188 - 26 Jan 2025
Viewed by 1450
Abstract
p27Kip1 is a key cell cycle gatekeeper governing the timing of Cyclin-dependent kinase (CDK) activation/inactivation and, consequently, cell proliferation. Structurally, the protein is largely unfolded, a feature that strongly increases its plasticity and interactors and enhances the number of regulated cellular processes. [...] Read more.
p27Kip1 is a key cell cycle gatekeeper governing the timing of Cyclin-dependent kinase (CDK) activation/inactivation and, consequently, cell proliferation. Structurally, the protein is largely unfolded, a feature that strongly increases its plasticity and interactors and enhances the number of regulated cellular processes. p27Kip1, like other intrinsically unstructured proteins, is post-translationally modified on several residues. These modifications affect its cellular localization and address p27Kip1 for specific interactions/functions. Several germline or somatic CDKN1B (the p27Kip1 encoding gene) mutations have been demonstrated to be associated with multiple endocrine neoplasia type 4 (MEN4), hairy cell leukemia, small-intestine neuroendocrine tumors, and breast and prostate cancers. Here, we analyzed the effect of four CDKN1B missense and nonsense mutations found in patients affected by MEN4 or cancers, namely, c.349C>T, p.P117S; c.397C>A, p.P133T; c.487C>T, p.Q163*; and c.511G>T, p.E171*. By transfecting breast cancer cell lines, we observed increased growth and cell motility for all the investigated mutants compared to wild-type p27Kip1 transfected cells. Furthermore, we discovered that the mutant forms exhibited altered phosphorylation on key residues and different localization or degradation mechanisms in comparison to the wild-type protein and suggested a possible region as crucial for the lysosome-dependent degradation of the protein. Finally, the loss of p27Kip1 ability in blocking cell proliferation was in part explained through the different binding efficiency that mutant p27Kip1 forms exhibited with Cyclin/Cyclin-dependent Kinase complexes (or proteins involved indirectly in that binding) with respect to the WT. Full article
Show Figures

Figure 1

13 pages, 1607 KiB  
Article
Clinical and Genomic Features of Androgen Indifferent Prostate Cancer
by Jack Masur, Aakrosh Ratan, Krzysztof Wierbilowicz, Adanma Ayanambakkam, Michelle L. Churchman, Laura S. Graham, George Daniel Grass, Sumati Gupta, Sean Q. Kern, Jennifer King, Zin Myint, Robert J. Rounbehler, Bodour Salhia, Eric A. Singer, Yousef Zakharia, Bryce M. Paschal and Paul V. Viscuse
Int. J. Mol. Sci. 2025, 26(2), 679; https://doi.org/10.3390/ijms26020679 - 15 Jan 2025
Viewed by 1832
Abstract
Androgen-indifferent prostate cancer (AIPC) is increasingly common and particularly lethal. Data describing these tumors are sparse, and AIPC remains a poorly understood malignancy. Utilizing the Oncology Research Information Exchange Network (ORIEN) database, we enriched for tumors with features of AIPC using previously described [...] Read more.
Androgen-indifferent prostate cancer (AIPC) is increasingly common and particularly lethal. Data describing these tumors are sparse, and AIPC remains a poorly understood malignancy. Utilizing the Oncology Research Information Exchange Network (ORIEN) database, we enriched for tumors with features of AIPC using previously described characteristics. Our AIPC cohort included three subgroups: aggressive variant prostate cancer (AVPC), neuroendocrine PC (NEPC), and double-negative PC (DNPC). Of 1496 total PC patients available for analysis, we identified 323 (22%) as MCRPC. Of those, 39 (12%) met AIPC criteria (17 AVPC, 13 NEPC, 9 DNPC) and 284 (88%) were non-AIPC. Forty-three percent of AIPC patients had de novo metastatic disease vs. 15% for non-AIPC (p = 0.003). Homologous recombination deficiency (HRD) and tumor mutational burden (TMB) did not differ between cohorts, but microsatellite instability scores (MSI) were significantly higher in AIPC (p = 0.019). Using Gene Set Enrichment Analysis (GSEA), we found that genes defining response to androgens and genes involved in oxidative phosphorylation were the most downregulated, whereas genes involved in epithelial–mesenchymal transition (EMT) and immune signaling were significantly upregulated in AIPC vs. non-AIPC. Our study demonstrates the potential for predefined criteria that aim to enrich for AIPC and suggests opportunities for therapeutic investigation. Full article
(This article belongs to the Special Issue Prostate Cancer: Novel Research and Innovative Therapeutic Strategies)
Show Figures

Figure 1

26 pages, 2549 KiB  
Review
Therapeutic Exploitation of Neuroendocrine Transdifferentiation Drivers in Prostate Cancer
by Zoe R. Maylin, Christopher Smith, Adam Classen, Mohammad Asim, Hardev Pandha and Yuzhuo Wang
Cells 2024, 13(23), 1999; https://doi.org/10.3390/cells13231999 - 3 Dec 2024
Cited by 1 | Viewed by 2588
Abstract
Neuroendocrine prostate cancer (NEPC), an aggressive and lethal subtype of prostate cancer (PCa), often arises as a resistance mechanism in patients undergoing hormone therapy for prostate adenocarcinoma. NEPC is associated with a significantly poor prognosis and shorter overall survival compared to conventional prostate [...] Read more.
Neuroendocrine prostate cancer (NEPC), an aggressive and lethal subtype of prostate cancer (PCa), often arises as a resistance mechanism in patients undergoing hormone therapy for prostate adenocarcinoma. NEPC is associated with a significantly poor prognosis and shorter overall survival compared to conventional prostate adenocarcinoma due to its aggressive nature and limited response to standard of care therapies. This transdifferentiation, or lineage reprogramming, to NEPC is characterised by the loss of androgen receptor (AR) and prostate-specific antigen (PSA) expression, and the upregulation of neuroendocrine (NE) biomarkers such as neuron-specific enolase (NSE), chromogranin-A (CHGA), synaptophysin (SYP), and neural cell adhesion molecule 1 (NCAM1/CD56), which are critical for NEPC diagnosis. The loss of AR expression culminates in resistance to standard of care PCa therapies, such as androgen-deprivation therapy (ADT) which target the AR signalling axis. This review explores the drivers of NE transdifferentiation. Key genetic alterations, including those in the tumour suppressor genes RB1, TP53, and PTEN, and changes in epigenetic regulators, particularly involving EZH2 and cell-fate-determining transcription factors (TFs) such as SOX2, play significant roles in promoting NE transdifferentiation and facilitate the lineage switch from prostate adenocarcinoma to NEPC. The recent identification of several other key novel drivers of NE transdifferentiation, including MYCN, ASCL1, BRN2, ONECUT2, and FOXA2, further elucidates the complex regulatory networks and pathways involved in this process. We suggest that, given the multifactorial nature of NEPC, novel therapeutic strategies that combine multiple modalities are essential to overcome therapeutic resistance and improve patient outcomes. Full article
Show Figures

Figure 1

18 pages, 19195 KiB  
Article
Unmasking Neuroendocrine Prostate Cancer with a Machine Learning-Driven Seven-Gene Stemness Signature That Predicts Progression
by Agustina Sabater, Pablo Sanchis, Rocio Seniuk, Gaston Pascual, Nicolas Anselmino, Daniel F. Alonso, Federico Cayol, Elba Vazquez, Marcelo Marti, Javier Cotignola, Ayelen Toro, Estefania Labanca, Juan Bizzotto and Geraldine Gueron
Int. J. Mol. Sci. 2024, 25(21), 11356; https://doi.org/10.3390/ijms252111356 - 22 Oct 2024
Cited by 1 | Viewed by 2867
Abstract
Prostate cancer (PCa) poses a significant global health challenge, particularly due to its progression into aggressive forms like neuroendocrine prostate cancer (NEPC). This study developed and validated a stemness-associated gene signature using advanced machine learning techniques, including Random Forest and Lasso regression, applied [...] Read more.
Prostate cancer (PCa) poses a significant global health challenge, particularly due to its progression into aggressive forms like neuroendocrine prostate cancer (NEPC). This study developed and validated a stemness-associated gene signature using advanced machine learning techniques, including Random Forest and Lasso regression, applied to large-scale transcriptomic datasets. The resulting seven-gene signature (KMT5C, DPP4, TYMS, CDC25B, IRF5, MEN1, and DNMT3B) was validated across independent cohorts and patient-derived xenograft (PDX) models. This signature demonstrated strong prognostic value for progression-free, disease-free, relapse-free, metastasis-free, and overall survival. Importantly, the signature not only identified specific NEPC subtypes, such as large-cell neuroendocrine carcinoma, which is associated with very poor outcomes, but also predicted a poor prognosis for PCa cases that exhibit this molecular signature, even when they were not histopathologically classified as NEPC. This dual prognostic and classifier capability makes the seven-gene signature a robust tool for personalized medicine, providing a valuable resource for predicting disease progression and guiding treatment strategies in PCa management. Full article
Show Figures

Figure 1

Back to TopTop