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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,381)

Search Parameters:
Keywords = brain cancers

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
34 pages, 1483 KB  
Review
Choice of Animal Models to Investigate Cell Migration and Invasion in Glioblastoma
by Piyanka Hettiarachchi and Taeju Park
Cancers 2025, 17(17), 2776; https://doi.org/10.3390/cancers17172776 - 26 Aug 2025
Abstract
Glioblastoma is an aggressive and prevalent form of brain cancer characterized by rapid tumor cell migration and invasion into surrounding healthy tissues, making it resistant to conventional treatments. Despite advances in therapeutic approaches, patient prognosis remains poor, with a median survival of approximately [...] Read more.
Glioblastoma is an aggressive and prevalent form of brain cancer characterized by rapid tumor cell migration and invasion into surrounding healthy tissues, making it resistant to conventional treatments. Despite advances in therapeutic approaches, patient prognosis remains poor, with a median survival of approximately 15 months. Tumor cell infiltration along perivascular spaces and white matter tracts is a major driver of recurrence, underscoring the need for experimental models that accurately capture these invasive behaviors. Animal models remain indispensable for this purpose, offering insights that cannot be fully replicated in vitro. This review focuses on applying animal models to elucidate the mechanisms underlying glioblastoma cell migration and invasion, which remain critical to improving therapeutic outcomes. By comparing the advantages of animal models with in vitro systems, we highlight the unique insights animal models provide, particularly in capturing the intricate dynamics of tumor cell motility. In particular, patient-derived xenograft (PDX) models preserve patient-specific heterogeneity and invasion patterns, such as white matter tract and perivascular infiltration, enabling clinically relevant drug testing. Zebrafish xenografts provide real-time, high-resolution visualization of tumor-vascular interactions, facilitating rapid assessment of invasion dynamics and early-stage drug screening. Genetically engineered models (GEM) allow precise discrimination of how defined genetic alterations drive specific invasive routes in the brain. Furthermore, we explore the use of advanced imaging techniques in these models to monitor tumor progression in real time. Moreover, we discuss the major drawbacks of these animal models, such as incomplete immune components and tumor microenvironment recapitulation. Ultimately, animal models are essential for bridging the gap between basic research and clinical application, offering a powerful platform for developing targeted strategies to combat glioblastoma’s relentless progression. Full article
(This article belongs to the Special Issue Cell Biology of Cancer Invasion: 2nd Edition)
Show Figures

Figure 1

21 pages, 1863 KB  
Article
Beyond Analgesia: Psychobiotics as an Adjunctive Approach to Pain Management in Gastrointestinal Oncology—A Post Hoc Analysis from the ProDeCa Study
by Georgios Tzikos, Alexandra-Eleftheria Menni, Helen Theodorou, Eleni Chamalidou, Ioannis M. Theodorou, George Stavrou, Anne D. Shrewsbury, Aikaterini Amaniti, Anastasia Konsta, Joulia K. Tsetis, Vasileios Grosomanidis and Katerina Kotzampassi
Nutrients 2025, 17(17), 2751; https://doi.org/10.3390/nu17172751 - 25 Aug 2025
Abstract
Background: Pain is a multifaceted and debilitating symptom in patients with gastrointestinal cancer, especially those undergoing surgical resection followed by chemotherapy. The interplay between inflammatory, neuropathic, and psychosocial components often renders conventional analgesia insufficient. Psychobiotics—probiotic strains with neuroactive properties—have recently emerged as [...] Read more.
Background: Pain is a multifaceted and debilitating symptom in patients with gastrointestinal cancer, especially those undergoing surgical resection followed by chemotherapy. The interplay between inflammatory, neuropathic, and psychosocial components often renders conventional analgesia insufficient. Psychobiotics—probiotic strains with neuroactive properties—have recently emerged as potential modulators of pain perception through neuroimmune and gut–brain axis pathways. Methods: This post hoc analysis is based on the ProDeCa randomized, placebo-controlled trial, which originally aimed to assess the psychotropic effects of a four-strain psychobiotic formulation in postoperative gastrointestinal cancer patients receiving chemotherapy. In the current analysis, we evaluated changes in pain perception among non-depressed and depressed participants, who received either psychobiotics or placebo, along with standard analgesic regimes. Pain was assessed at baseline, after a month of treatment, and at follow-up, 2 months thereafter, using the Short-Form McGill Pain Questionnaire (SF-MPQ), capturing both sensory and affective components, as well as with the Present Pain Intensity and the VAS scores. Results: Psychobiotic-treated participants—particularly the non-depressed ones—exhibited a significant reduction in both quantitative and qualitative pain indices over time compared with placebo-treated ones. Improvements were noted in total pain rating index scores, sensory and affective subscales, and present pain intensity. These effects were sustained up to 2 months after intervention. In contrast, placebo groups demonstrated worsening in pain scores, probably influenced by ongoing chemotherapy and disease progression. The analgesic effect was less pronounced but still observable in the subgroup with symptoms of depression. Conclusions: Adjunctive psychobiotic therapy appears to beneficially modulate pain perception in gastrointestinal oncology patients receiving chemotherapy, with the most pronounced effects being in non-depressed individuals. These findings suggest psychobiotics as a promising non-opioid add-on for comprehensive cancer pain management and support further investigation in larger pain-targeted trials. Full article
Show Figures

Figure 1

16 pages, 533 KB  
Perspective
The Future of Oncology in Psychiatric Medications
by Napoleon Waszkiewicz
J. Clin. Med. 2025, 14(17), 6003; https://doi.org/10.3390/jcm14176003 - 25 Aug 2025
Abstract
Recent years have provided numerous reports on the mechanisms of action of psychiatric medications (antidepressants, antipsychotics, mood stabilizers, and antidementia drugs) that directly inhibit the growth of cancer cells, as well as on their indirect effects on the psyche and immune system, and [...] Read more.
Recent years have provided numerous reports on the mechanisms of action of psychiatric medications (antidepressants, antipsychotics, mood stabilizers, and antidementia drugs) that directly inhibit the growth of cancer cells, as well as on their indirect effects on the psyche and immune system, and their supportive effects on chemotherapeutic agents. The mechanisms of the anticancer activity of psychiatric drugs include inhibition of dopamine and N-methyl-D-aspartate receptors that work via signaling pathways (PI3K/AKT/mTOR/NF-κB, ERK, Wnt/ß-catenin, and bcl2), metabolic pathways (ornithine decarboxylase, intracellular cholesterol transport, lysosomal enzymes, and glycolysis), autophagy, Ca2+-dependent signaling cascades, and various other proteins (actin-related protein complex, sirtuin 1, p21, p53, etc.). The anticancer potential of psychiatric drugs seems to be extremely broad, and the most extensive anticancer literature has been reported on antidepressants (fluoxetine, amitriptyline, imipramine, mirtazapine, and St John’s Wort) and antipsychotics (chlorpromazine, pimozide, thioridazine, and trifluoperazine). Among mood stabilizers, lithium and valproates have the largest body of literature. Among antidementia drugs, memantine has documented anticancer effects, while there is limited evidence for galantamine. Of the new psychiatric substances, the antipsychotic drug brexpiprazole and the antidepressant vortioxetine have a very interesting body of literature regarding glioblastoma, based on in vitro and in vivo animal survival studies. Their use in brain tumors and metastases is particularly compelling, as these substances readily cross the blood–brain barrier (BBB). Moreover, the synergistic effect of psychiatric drugs with traditional cancer treatment seems to be extremely important in the fight against chemo- and radio-resistance of tumors. Although there are some studies describing the possible carcinogenic effects of psychiatric drugs in animals, the anticancer effect seems to be extremely significant, especially in combination treatment with radio/chemotherapy. The emerging evidence supporting the anticancer properties of psychiatric drugs presents an exciting frontier in oncology. The anticancer properties of psychiatric drugs may prove particularly useful in the period between chemotherapy and radiotherapy sessions to maintain the tumor-inhibitory effect. While further research is necessary to elucidate the mechanisms, clinical implications, dose-dependence of the effect, and clear guidelines for the use of psychiatric medications in cancer therapy, the potential for these commonly prescribed medications to contribute to cancer treatment enhances their value in the management of patients facing the dual challenges of mental health and cancer. Full article
(This article belongs to the Section Mental Health)
Show Figures

Figure 1

20 pages, 347 KB  
Article
Site-Specific Inflammatory Signatures in Metastatic NSCLC: Insights from Routine Blood Count Parameters
by Vlad-Norin Vornicu, Alina-Gabriela Negru, Razvan Constantin Vonica, Andrei Alexandru Cosma, Sorin Saftescu, Mihaela Maria Pasca-Fenesan and Anca Maria Cimpean
Medicina 2025, 61(9), 1521; https://doi.org/10.3390/medicina61091521 - 25 Aug 2025
Abstract
Background and Objectives: Systemic inflammatory markers from an ordinary complete blood count (CBC) may foreshadow where non-small-cell lung cancer (NSCLC) will first spread, but organ-specific signatures remain poorly defined. Materials and Methods: We retrospectively reviewed 302 adults (mean age 60.7 ± [...] Read more.
Background and Objectives: Systemic inflammatory markers from an ordinary complete blood count (CBC) may foreshadow where non-small-cell lung cancer (NSCLC) will first spread, but organ-specific signatures remain poorly defined. Materials and Methods: We retrospectively reviewed 302 adults (mean age 60.7 ± 13.4 years; 80.8% men) with stage IV NSCLC managed at OncoHelp Medical Center, Timișoara, between January 2022 and December 2024. Eligibility demanded a single radiologically confirmed distant site at diagnosis and pre-treatment CBC. Neutrophil-to-lymphocyte (NLR), platelet-to-lymphocyte (PLR), and lymphocyte-to-monocyte (LMR) ratios were compared across pleural (n = 52), bone (n = 86), liver (n = 66), and brain (n = 98) metastases using Kruskal–Wallis tests with Bonferroni adjustment; z-standardized logistic models identified independent predictors. Results: Metastases clustered most often in brain (32.5%), followed by bone (28.5%), liver (21.9%), and pleura (17.2%). Median PLR rose selectively in pleural disease (274 vs. 217–253 in other sites; p = 0.006). LMR fell to 2.0 in bone but climbed to 2.8 in brain lesions (p = 0.032 and 0.008, respectively). NLR was globally elevated (6.7–7.6), yet differed significantly only for bone and liver deposits. Logistic modeling showed that each standard-deviation rise in absolute neutrophil count quadrupled the odds of hepatic involvement (Odd Ratio (OR) 4.26; 99% Confidence inerval (CI) 2.20–6.25), monocytosis nearly doubled bone risk (OR 1.83; 1.01–3.33), while higher erythrocytes, eosinophils, and lymphocytes independently protected against pleural seeding (all p < 0.01). Age-stratified analysis revealed that osseous and cerebral metastases predominated in patients ≤ 50 years, whereas inflammatory indices were age-invariant. Conclusions: Routine CBC ratios encode distinct “inflammatory fingerprints” that mirror the first metastatic destination in NSCLC: platelets herald pleural spread, neutrophils favor liver and bone, and divergent lymphocyte–monocyte balances separate bone from brain. Although no substitute for cross-sectional imaging, these low-cost markers could refine clinical suspicion, guide targeted work-up, and illuminate the biology of organ-selective dissemination, particularly in resource-limited settings. Full article
(This article belongs to the Special Issue Insights and Advances in Cancer Biomarkers)
Show Figures

Figure 1

14 pages, 5518 KB  
Article
NOD2 Promotes Glioblastoma Progression Through Effects on Epithelial–Mesenchymal Transition and Cancer Stemness
by Eshrat Jahan, Shubhash Chandra Chaudhary, S M Abdus Salam, Eun-Jung Ahn, Nah Ihm Kim, Tae-Young Jung, Jong-Hwan Park, Sung Sun Kim, Ji Young Lee, Kyung-Hwa Lee and Kyung-Sub Moon
Biomedicines 2025, 13(8), 2041; https://doi.org/10.3390/biomedicines13082041 - 21 Aug 2025
Viewed by 499
Abstract
Background: Glioblastoma multiforme (GBM) represents one of the most aggressive and lethal primary brain malignancies, characterized by rapid proliferation, extensive invasiveness, and a dismal prognosis. Emerging evidence implicates nucleotide-binding oligomerization domain-containing protein 2 (NOD2), an intracellular pattern recognition receptor, as [...] Read more.
Background: Glioblastoma multiforme (GBM) represents one of the most aggressive and lethal primary brain malignancies, characterized by rapid proliferation, extensive invasiveness, and a dismal prognosis. Emerging evidence implicates nucleotide-binding oligomerization domain-containing protein 2 (NOD2), an intracellular pattern recognition receptor, as a potential driver of GBM progression. This study investigates NOD2’s role in promoting glioblastoma through its effects on the epithelial–mesenchymal transition (EMT) and cancer stem cell (CSC) markers. Methods: NOD2 expression levels and survival outcomes were assessed using TCGA data from GBM tumor samples (n = 153) and normal brain tissues (n = 5). NOD2 protein expression was validated in glioma cell lines using Western blot and immunofluorescence analyses. Functional studies employed siRNA-mediated NOD2 knockdown to evaluate effects on cellular proliferation, migration, invasion, and colony formation, while correlations between NOD2 and EMT/CSC markers were assessed. Results: The analysis of TCGA data revealed a significantly elevated NOD2 expression in GBM tumors compared to normal brain tissue, with a high NOD2 expression correlating with a reduced disease-free survival in GBM patients. All tested glioma cell lines demonstrated robust NOD2 expression. Functional analyses demonstrated that NOD2 depletion substantially impaired cellular proliferation, migration, invasion, and the colony-forming capacity. Mechanistically, siRNA-mediated NOD2 knockdown significantly decreased the expression of EMT (Snail, SLUG, Vimentin) and CSC markers (CD44, CD133) at both protein and mRNA levels. Conclusions: Our results indicate that NOD2 contributes to GBM progression by influencing EMT and CSC pathways. These findings suggest NOD2’s potential as a therapeutic target in glioblastoma, highlighting the need for further mechanistic studies and therapeutic exploration. Full article
(This article belongs to the Special Issue Mechanisms and Novel Therapeutic Approaches for Gliomas)
Show Figures

Figure 1

14 pages, 8079 KB  
Article
Epilepsy Associated Gene, Pcdh7, Is Dispensable for Brain Development in Mice
by Jennifer Rakotomamonjy, Devin Davies, Xavier Valencia, Olivia Son, Ximena Gomez-Maqueo and Alicia Guemez-Gamboa
Genes 2025, 16(8), 985; https://doi.org/10.3390/genes16080985 - 21 Aug 2025
Viewed by 228
Abstract
Background/Objectives: Protocadherin 7 (Pcdh7) belongs to the protocadherin family, the largest subgroup of cell adhesion molecules. Members of this family are highly expressed in the brain, where they serve fundamental roles in many neurodevelopmental processes, including axon guidance, dendrite self-avoidance, [...] Read more.
Background/Objectives: Protocadherin 7 (Pcdh7) belongs to the protocadherin family, the largest subgroup of cell adhesion molecules. Members of this family are highly expressed in the brain, where they serve fundamental roles in many neurodevelopmental processes, including axon guidance, dendrite self-avoidance, and synaptic formation. PCDH7 has been strongly associated with epilepsy in multiple genome-wide association studies (GWAS), as well as with schizophrenia, PTSD, and childhood aggression. Despite these associations, the specific contributions of PCDH7 to epileptogenesis and brain development remain largely unexplored. Most of the existing literature on PCDH7 focuses on its function during cancer progression, with only one study suggesting that PCDH7 regulates dendritic spine morphology and synaptic function via interaction with GluN1. Methods: Here, we generate, validate, and characterize a murine null Pcdh7 allele in which a large deletion was introduced by CRISPR. Results: Analysis of embryonic, postnatal, and adult brain datasets confirmed PCDH7 widespread expression. Pcdh7+/− and Pcdh7−/− mice present no gross morphological defects and normal cortical layer formation. However, a seizure susceptibility assay revealed increased latencies in Pcdh7+/− mice, but not in Pcdh7+/+ and Pcdh7−/− mice, potentially explaining the association of PCDH7 with epilepsy. Conclusions: This initial characterization of Pcdh7 null mice suggests that, despite its widespread expression in the CNS and involvement in human epilepsy, PCDH7 is not essential for murine brain development and thus is not a suitable animal model for understanding PCDH7 disruption in humans. However, further detailed analysis of this mouse model may reveal circuit or synaptic abnormalities in Pcdh7 null brains. Full article
(This article belongs to the Special Issue The Genetic and Epigenetic Basis of Neurodevelopmental Disorders)
Show Figures

Figure 1

38 pages, 2700 KB  
Review
From Microbial Switches to Metabolic Sensors: Rewiring the Gut–Brain Kynurenine Circuit
by Masaru Tanaka and László Vécsei
Biomedicines 2025, 13(8), 2020; https://doi.org/10.3390/biomedicines13082020 - 19 Aug 2025
Viewed by 335
Abstract
The kynurenine (KYN) metabolic pathway sits at the crossroads of immunity, metabolism, and neurobiology, yet its clinical translation remains fragmented. Emerging spatial omics, wearable chronobiology, and synthetic microbiota studies reveal that tryptophan (Trp) metabolism is regulated by distinct cellular “checkpoints” along the gut–brain [...] Read more.
The kynurenine (KYN) metabolic pathway sits at the crossroads of immunity, metabolism, and neurobiology, yet its clinical translation remains fragmented. Emerging spatial omics, wearable chronobiology, and synthetic microbiota studies reveal that tryptophan (Trp) metabolism is regulated by distinct cellular “checkpoints” along the gut–brain axis, finely modulated by sex differences, circadian rhythms, and microbiome composition. However, current interventions tackle single levers in isolation, leaving a key gap in the precision control of Trp’s fate. To address this, we drew upon an extensive body of the primary literature and databases, mapping enzyme expression across tissues at single-cell resolution and linking these profiles to clinical trials investigating dual indoleamine 2,3-dioxygenase 1 (IDO1)/tryptophan 2,3-dioxygenase (TDO) inhibitors, engineered probiotics, and chrono-modulated dosing strategies. We then developed decision-tree algorithms that rank therapeutic combinations against biomarker feedback loops derived from real-time saliva, plasma, and stool metabolomics. This synthesis pinpoints microglial and endothelial KYN hotspots, quantifies sex-specific chronotherapeutic windows, and identifies engineered Bifidobacterium consortia and dual inhibitors as synergistic nodes capable of reducing immunosuppressive KYN while preserving neuroprotective kynurenic acid. Here, we highlight a framework that couples lifestyle levers, bio-engineered microbes, and adaptive pharmaco-regimens into closed-loop “smart protocols.” By charting these intersections, this study offers a roadmap for biomarker-guided, multidisciplinary interventions that could recalibrate KYN metabolic activity across cancer, mood, neurodegeneration, and metabolic disorders, appealing to clinicians, bioengineers, and systems biologists alike. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Graphical abstract

15 pages, 1929 KB  
Article
Direct oHSV Infection Induces DC Maturation and a Tumor Therapeutic Response
by Doyeon Kim, Michael Kelly, Jack Hedberg, Alexia K. Martin, Ilse Hernandez-Aguirre, Yeaseul Kim, Lily R. Cain, Ravi Dhital and Kevin A. Cassady
Viruses 2025, 17(8), 1134; https://doi.org/10.3390/v17081134 - 19 Aug 2025
Viewed by 351
Abstract
Oncolytic herpes simplex virus (oHSV) is a promising cancer immunotherapy that induces tumor cell lysis and stimulates anti-tumor immunity. Our previous single-cell RNA sequencing analysis of oHSV-treated medulloblastoma tumors revealed expansion and activation of tumor-infiltrating dendritic cells (DCs), and direct oHSV infection of [...] Read more.
Oncolytic herpes simplex virus (oHSV) is a promising cancer immunotherapy that induces tumor cell lysis and stimulates anti-tumor immunity. Our previous single-cell RNA sequencing analysis of oHSV-treated medulloblastoma tumors revealed expansion and activation of tumor-infiltrating dendritic cells (DCs), and direct oHSV infection of DCs within the brain. While the therapeutic effects of oHSVs have been primarily attributed to tumor cell infection, we hypothesize that direct infection of DCs also contributes to therapeutic efficacy by promoting DC maturation and immune activation. Although the oHSV infection in DCs was abortive, it led to increased expression of major histocompatibility complex (MHC) class I/II and co-stimulatory molecules. oHSV-infected DCs activated naïve CD4+ and CD8+ T cells, inducing expression of CD69 and CD25. These primed T cells exhibited enhanced cytotoxicity against CT-2A glioma cells. Adoptive transfer of oHSV-infected DCs via subcutaneous injection near inguinal lymph nodes delayed tumor growth in a syngeneic CT-2A glioma model, independent of tumor viral replication and lysis. Mechanistically, our in vitro studies demonstrate that oHSV can directly infect and functionally activate DCs, enabling them to prime effective anti-tumor T cell responses. This study highlights the anti-tumor potential of leveraging oHSV-infected DCs to augment viroimmunotherapy as a cancer therapeutic. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

28 pages, 3851 KB  
Review
Technological Advances and Medical Applications of Implantable Electronic Devices: From the Heart, Brain, and Skin to Gastrointestinal Organs
by Jonghyun Lee, Sung Yong Han and Young Woo Kwon
Biosensors 2025, 15(8), 543; https://doi.org/10.3390/bios15080543 - 18 Aug 2025
Viewed by 469
Abstract
Implantable electronic devices are driving innovation in modern medical technology and have significantly improved patients’ quality of life. This review comprehensively analyzes the latest technological trends in implantable electronic devices used in major organs, including the heart, brain, and skin. Additionally, it explores [...] Read more.
Implantable electronic devices are driving innovation in modern medical technology and have significantly improved patients’ quality of life. This review comprehensively analyzes the latest technological trends in implantable electronic devices used in major organs, including the heart, brain, and skin. Additionally, it explores the potential for application in the gastrointestinal system, particularly in the field of biliary stents, in which development has been limited. In the cardiac field, wireless pacemakers, subcutaneous implantable cardioverter-defibrillators, and cardiac resynchronization therapy devices have been commercialized, significantly improving survival rates and quality of life of patients with cardiovascular diseases. In the field of brain–neural interfaces, biocompatible flexible electrodes and closed-loop deep brain stimulation have improved treatments of neurological disorders, such as Parkinson’s disease and epilepsy. Skin-implantable devices have revolutionized glucose management in patients with diabetes by integrating continuous glucose monitoring and automated insulin delivery systems. Future development of implantable electronic devices incorporating pressure or pH sensors into biliary stents in the gastrointestinal system may significantly improve the prognosis of patients with bile duct cancer. This review systematically organizes the technological advances and clinical outcomes in each field and provides a comprehensive understanding of implantable electronic devices by suggesting future research directions. Full article
(This article belongs to the Section Biosensors and Healthcare)
Show Figures

Figure 1

22 pages, 1030 KB  
Article
Current and Emerging Therapeutic Strategies for Limited- and Extensive-Stage Small-Cell Lung Cancer
by Walid Shalata, Rashad Naamneh, Wenad Najjar, Mohnnad Asla, Adam Abu Gameh, Mahmoud Abu Amna, Leonard Saiegh and Abed Agbarya
Med. Sci. 2025, 13(3), 142; https://doi.org/10.3390/medsci13030142 - 18 Aug 2025
Viewed by 567
Abstract
Background: Small-cell lung cancer (SCLC) is a highly aggressive neuroendocrine malignancy characterized by rapid growth, early metastatic dissemination, and a dismal prognosis. For decades, treatment paradigms remained largely stagnant, particularly for extensive-stage disease (ES-SCLC). However, the last five years have witnessed a significant [...] Read more.
Background: Small-cell lung cancer (SCLC) is a highly aggressive neuroendocrine malignancy characterized by rapid growth, early metastatic dissemination, and a dismal prognosis. For decades, treatment paradigms remained largely stagnant, particularly for extensive-stage disease (ES-SCLC). However, the last five years have witnessed a significant evolution in the therapeutic landscape. Methods: The information for this article was gathered by synthesizing data from several key sources. This article synthesizes the evidence supporting current standards of care for both limited-stage (LS-SCLC) and ES-SCLC, incorporating data from pivotal clinical trials, a network meta-analysis of first-line chemoimmunotherapy regimens, and a critical appraisal of international treatment guidelines, and a critical analysis of international treatment guidelines from prominent organizations like the National Comprehensive Cancer Network (NCCN) and the European Society for Medical Oncology (ESMO). This comprehensive approach allows for a robust and well-supported summary of the current therapeutic landscape. Results: For limited-stage SCLC (LS-SCLC), concurrent chemoradiotherapy (cCRT) remains the curative-intent standard, but its efficacy is now being augmented by consolidative immunotherapy, as demonstrated by the landmark ADRIATIC trial. The role of prophylactic cranial irradiation (PCI) in LS-SCLC is being re-evaluated in the era of high-sensitivity brain imaging and concerns over neurotoxicity. For ES-SCLC, the treatment paradigm has been fundamentally transformed by the integration of immune checkpoint inhibitors (ICIs) with platinum–etoposide chemotherapy, establishing a new standard of care that offers a modest but consistent survival benefit. Conclusions: The treatment of SCLC has been significantly advanced by the integration of immunotherapy, particularly for extensive-stage disease, which has established a new standard of care and improved patient outcomes. Looking to the future, the quest for predictive biomarkers and the development of novel therapeutic classes, such as Bi-specific T-cell Engagers (BiTEs) and antibody–drug conjugates, promise to build upon recent progress and offer new hope for improving the dismal prognosis associated with this disease. Full article
(This article belongs to the Special Issue Feature Papers in Section Cancer and Cancer-Related Diseases)
Show Figures

Figure 1

17 pages, 258 KB  
Article
Survival Patterns and Mortality Causes in Patients with Invasive Ependymoma: A Retrospective Cohort Analysis from 2000 to 2019
by Anas Elgenidy, Khaled Saad, Amir Aboelgheet, Eman F. Gad, Usama El-Shokhaiby, Thamer A. M. Alruwaili, Abdelrahman N. Abdelal, Kawashty R. Mohamed, Mohammad Bazzazeh, Mohamed Hesn, Abdelrahman H. Elshimy, Aya Sayed, Sara Magdy, Doaa Ali Gamal, Amira A. Elhoufey, Shymaa Adel Ismael, Ahmed M. Afifi, Mohamed Fahmy M. Ibrahim and Amany Ragab
Med. Sci. 2025, 13(3), 139; https://doi.org/10.3390/medsci13030139 - 16 Aug 2025
Viewed by 278
Abstract
Background: Ependymomas are primary CNS neoplasms that arise from the ependymal cells of the brain and spinal cord, accounting for 3–6% of all CNS tumors. Aims: This study provides a comprehensive analysis of ependymoma survival patterns and examines non-cancer causes of death in [...] Read more.
Background: Ependymomas are primary CNS neoplasms that arise from the ependymal cells of the brain and spinal cord, accounting for 3–6% of all CNS tumors. Aims: This study provides a comprehensive analysis of ependymoma survival patterns and examines non-cancer causes of death in the US. Methods: This retrospective study used data from SEER 17 registries between 2000 and 2019 to evaluate the incidence of ependymoma, as well as the survival and mortality trends in the US. Results: A total of 3821 patients were included, with 842 (22%) deaths. The highest mortality was observed in younger patients (<18 years) within one year of diagnosis (SMR, 54.77; 95% CI, 38.95–74.88). Brain and other nervous system cancers were the leading causes of death, followed by non-cancer causes, particularly cerebrovascular diseases, pneumonia, influenza, and septicemia. The survival rates observed at one, three, and five years were 94% (95% CI: 0.94–0.95), 88% (95% CI: 0.87–0.89), and 84% (95% CI: 0.82–0.85), respectively. Conditional survival improved over time, with a three-year conditional relative survival rate of 92% after one year of diagnosis and 96% for those who survived five years. Conclusion: The death rate was highest among pediatric patients under 18 years of age. Cerebrovascular disorders were the leading non-cancer cause of death across all time intervals. The probability of surviving for three years increases for patients who have already survived one, three, or five years post-diagnosis. Full article
(This article belongs to the Section Cancer and Cancer-Related Research)
54 pages, 9063 KB  
Article
Cell Settling, Migration, and Stochastic Cancer Gene Expression Suggest Potassium Membrane Flux May Initiate pH Reversal
by Marie E. Beckner
Biomolecules 2025, 15(8), 1177; https://doi.org/10.3390/biom15081177 - 16 Aug 2025
Viewed by 511
Abstract
Attraction of glioblastoma cells to potassium was suspected when glioblastoma cells clustered around dying cells and migrated towards serum (high [K+]) and increased potassium. Potassium channel proteins (KCN family, 90 members) mediating alterations in the transmembrane flux may provide K+ [...] Read more.
Attraction of glioblastoma cells to potassium was suspected when glioblastoma cells clustered around dying cells and migrated towards serum (high [K+]) and increased potassium. Potassium channel proteins (KCN family, 90 members) mediating alterations in the transmembrane flux may provide K+ that releases H+ bound to inner membranes in cancer cells for cytosolic proton transfer, possibly conformational in water (Grotthuss), to extrusion sites. Cell settling and migration assay results led to collecting 70 studies, unbiased by the authors for inclusion of KCN genes, that detected KCN differentially expressed genes (DEGs). Of 53 KCN DEGs found among 29 malignancies, 62.3% encoded H+-sensitive proteins. KCN DEGs encoding H+-sensitive proteins were more prevalent in 50 studies involving one or more categories (seven oncogenes and histone/DNA modifiers) versus those with none; p = 0.0325. Pertinent genes for lactate outflow, etc., had relatively normal levels of expression. Brain tumors in REMBRANDT (database) showed altered expression of KCN genes encoding H+-sensitive proteins in glioblastomas versus less invasive oligodendrogliomas of patients on anti-seizure medications, with less KCNJ16/Kir5.1; p = 5.32 × 10−8 in glioblastomas. Altered H+-sensitive potassium flux via the KCN family, downstream of oncogenes and histone/DNA modifiers, putatively incites proton transfers for H+ release during pH reversal (pHi > pHe) in cancer. Full article
(This article belongs to the Collection Feature Papers in Chemical Biology)
Show Figures

Graphical abstract

37 pages, 9132 KB  
Perspective
The Evidence That Brain Cancers Could Be Effectively Treated with In-Home Radiofrequency Waves
by Gary W. Arendash
Cancers 2025, 17(16), 2665; https://doi.org/10.3390/cancers17162665 - 15 Aug 2025
Viewed by 469
Abstract
There is currently no effective therapeutic capable of arresting or inducing regression of primary or metastatic brain cancers. This article presents both pre-clinical and clinical studies supportive that a new bioengineered technology could induce regression and/or elimination of primary and metastatic brain cancers [...] Read more.
There is currently no effective therapeutic capable of arresting or inducing regression of primary or metastatic brain cancers. This article presents both pre-clinical and clinical studies supportive that a new bioengineered technology could induce regression and/or elimination of primary and metastatic brain cancers through three disease-modifying mechanisms. Transcranial Radiofrequency Wave Treatment (TRFT) is non-thermal, non-invasive and self-administered in-home to safely provide radiofrequency waves to the entire human brain. Since TRFT has already been shown to stop and reverse the cognitive decline of Alzheimer’s Disease in small studies, evidence is provided that three key mechanisms of TRFT action, alone or in synergy, could effectively treat brain cancers: (1) enhancement of brain meningeal lymph flow to increase immune trafficking between the brain cancer and cervical lymph nodes, resulting in a robust immune attack on the brain cancer; (2) rebalancing of the immune system’s cytokines within the brain or brain cancer environment to decrease inflammation therein and thus make for an inhospitable environment for brain cancer growth; (3) direct anti-proliferation/antigrowth affects within the brain tumor microenvironment. Importantly, these mechanisms of TRFT action could be effective against both visualized brain tumors and those that are yet too small to be identified through brain imaging. The existing animal and human clinical evidence presented in this perspective article justifies TRFT to be clinically tested immediately against both primary and metastatic brain cancers as monotherapy or possibly in combination with immune checkpoint inhibitors. Full article
(This article belongs to the Special Issue Emerging Research on Primary Brain Tumors)
Show Figures

Figure 1

26 pages, 2099 KB  
Review
Cannabis Medicine 2.0: Nanotechnology-Based Delivery Systems for Synthetic and Chemically Modified Cannabinoids for Enhanced Therapeutic Performance
by Izabela Żółnowska, Aleksandra Gostyńska-Stawna, Anna Jelińska and Maciej Stawny
Nanomaterials 2025, 15(16), 1260; https://doi.org/10.3390/nano15161260 - 15 Aug 2025
Viewed by 329
Abstract
The therapeutic potential of cannabinoids and other ligands of cannabinoid receptors attracts considerable attention due to their diverse pharmacological effects and utility in various medical applications. However, challenges such as low solubility, limited bioavailability, and potential side effects hinder their broad clinical use. [...] Read more.
The therapeutic potential of cannabinoids and other ligands of cannabinoid receptors attracts considerable attention due to their diverse pharmacological effects and utility in various medical applications. However, challenges such as low solubility, limited bioavailability, and potential side effects hinder their broad clinical use. Nanoformulation techniques offer a promising approach to address these issues and optimize the therapeutic effectiveness of cannabinoids and other cannabinoid receptor ligands. This comprehensive review explores the advancements in nanoformulation strategies to enhance the therapeutic efficacy and safety of synthetic cannabinoids and related compounds, such as CB13, rimonabant, and HU-211, which have been studied in a range of preclinical models addressing conditions such as neuropathic pain, depression, and cancer. The review discusses various nanocarriers employed in this field, including lipid-based, polymeric, and hybrid nanoparticles, micelles, emulsions, and other nanoengineered carriers. In addition to formulation approaches, this review provides an in-depth analysis of chemical structures and their effect on compound activity, especially in the context of the affinity for the cannabinoid type 1 receptor in the brain, which is chiefly responsible for the psychoactive effects. The provided summary of research concerning either chemical modifications of existing cannabinoids or the creation of new compounds that interact with cannabinoid receptors, followed by the development of nanoformulations for these agents, allows for the identification of new research directions and future perspectives for Cannabis-based medicine. In conclusion, the combination of nanotechnology and cannabinoid pharmacology holds promise for delivering more effective and safer therapeutic solutions for a broad spectrum of medical conditions, making this an exciting area of research with profound implications for the healthcare and pharmaceutical industries. Full article
(This article belongs to the Section Nanofabrication and Nanomanufacturing)
Show Figures

Graphical abstract

24 pages, 1942 KB  
Review
The Pivotal Role of NF-κB in Glioblastoma: Mechanisms of Activation and Therapeutic Implications
by Vanajothi Ramar, Shanchun Guo, Guangdi Wang and Mingli Liu
Int. J. Mol. Sci. 2025, 26(16), 7883; https://doi.org/10.3390/ijms26167883 - 15 Aug 2025
Viewed by 263
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and lethal primary brain tumor in adults, characterized by high intratumoral heterogeneity, therapy resistance, and poor prognosis. Nuclear factor-κB (NF-κB) signaling plays a pivotal role in GBM pathogenesis by promoting proliferation, invasion, inflammation, immune evasion, and [...] Read more.
Glioblastoma multiforme (GBM) is the most aggressive and lethal primary brain tumor in adults, characterized by high intratumoral heterogeneity, therapy resistance, and poor prognosis. Nuclear factor-κB (NF-κB) signaling plays a pivotal role in GBM pathogenesis by promoting proliferation, invasion, inflammation, immune evasion, and treatment resistance. This review provides a comprehensive overview of canonical and non-canonical NF-κB signaling pathways and their molecular mechanisms in GBM, with a focus on their regulation in glioma stem-like cells (GSCs), interactions with key oncogenic factors (including STAT3, FOSL1, and TRPM7), and roles in maintaining tumor stemness, metabolic adaptation, and angiogenesis. We further discuss the reciprocal regulatory dynamics between NF-κB and non-coding RNAs (ncRNAs), particularly microRNAs, highlighting novel ncRNA-mediated epigenetic switches that shape GBM cell plasticity and subtype specification. Additionally, we examine the influence of NF-κB in modulating the tumor microenvironment (TME), where it orchestrates pro-tumorigenic cytokine production, immune cell reprogramming, and stromal remodeling. Finally, we review current NF-κB-targeting therapeutic strategies in GBM, including clinical trial data on small-molecule inhibitors and combinatorial approaches. Understanding the multifaceted roles of NF-κB in GBM offers new insights into targeted therapies aimed at disrupting tumor-promoting circuits within both cancer cells and the TME. Full article
(This article belongs to the Special Issue Future Perspectives and Challenges in Molecular Research of Glioma)
Show Figures

Figure 1

Back to TopTop