Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (39)

Search Parameters:
Keywords = refractory glioblastoma

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
50 pages, 937 KiB  
Review
Precision Neuro-Oncology in Glioblastoma: AI-Guided CRISPR Editing and Real-Time Multi-Omics for Genomic Brain Surgery
by Matei Șerban, Corneliu Toader and Răzvan-Adrian Covache-Busuioc
Int. J. Mol. Sci. 2025, 26(15), 7364; https://doi.org/10.3390/ijms26157364 - 30 Jul 2025
Viewed by 413
Abstract
Precision neurosurgery is rapidly evolving as a medical specialty by merging genomic medicine, multi-omics technologies, and artificial intelligence (AI) technology, while at the same time, society is shifting away from the traditional, anatomic model of care to consider a more precise, molecular model [...] Read more.
Precision neurosurgery is rapidly evolving as a medical specialty by merging genomic medicine, multi-omics technologies, and artificial intelligence (AI) technology, while at the same time, society is shifting away from the traditional, anatomic model of care to consider a more precise, molecular model of care. The general purpose of this review is to contemporaneously reflect on how these advances will impact neurosurgical care by providing us with more precise diagnostic and treatment pathways. We hope to provide a relevant review of the recent advances in genomics and multi-omics in the context of clinical practice and highlight their transformational opportunities in the existing models of care, where improved molecular insights can support improvements in clinical care. More specifically, we will highlight how genomic profiling, CRISPR-Cas9, and multi-omics platforms (genomics, transcriptomics, proteomics, and metabolomics) are increasing our understanding of central nervous system (CNS) disorders. Achievements obtained with transformational technologies such as single-cell RNA sequencing and intraoperative mass spectrometry are exemplary of the molecular diagnostic possibilities in real-time molecular diagnostics to enable a more directed approach in surgical options. We will also explore how identifying specific biomarkers (e.g., IDH mutations and MGMT promoter methylation) became a tipping point in the care of glioblastoma and allowed for the establishment of a new taxonomy of tumors that became applicable for surgeons, where a change in practice enjoined a different surgical resection approach and subsequently stratified the adjuvant therapies undertaken after surgery. Furthermore, we reflect on how the novel genomic characterization of mutations like DEPDC5 and SCN1A transformed the pre-surgery selection of surgical candidates for refractory epilepsy when conventional imaging did not define an epileptogenic zone, thus reducing resective surgery occurring in clinical practice. While we are atop the crest of an exciting wave of advances, we recognize that we also must be diligent about the challenges we must navigate to implement genomic medicine in neurosurgery—including ethical and technical challenges that could arise when genomic mutation-based therapies require the concurrent application of multi-omics data collection to be realized in practice for the benefit of patients, as well as the constraints from the blood–brain barrier. The primary challenges also relate to the possible gene privacy implications around genomic medicine and equitable access to technology-based alternative practice disrupting interventions. We hope the contribution from this review will not just be situational consolidation and integration of knowledge but also a stimulus for new lines of research and clinical practice. We also hope to stimulate mindful discussions about future possibilities for conscientious and sustainable progress in our evolution toward a genomic model of precision neurosurgery. In the spirit of providing a critical perspective, we hope that we are also adding to the larger opportunity to embed molecular precision into neuroscience care, striving to promote better practice and better outcomes for patients in a global sense. Full article
(This article belongs to the Special Issue Molecular Insights into Glioblastoma Pathogenesis and Therapeutics)
Show Figures

Figure 1

24 pages, 3012 KiB  
Article
Structural Activity Relationship Analysis of New Diphenyl PFI-3 Analogues Targeting for the Treatment of Glioblastoma
by Dong-Jin Hwang, Chuanhe Yang, Yinan Wang, Hannah Kelso, Satyanarayana Pochampally, Lawrence M. Pfeffer and Duane D. Miller
Pharmaceuticals 2025, 18(5), 608; https://doi.org/10.3390/ph18050608 - 23 Apr 2025
Cited by 1 | Viewed by 779
Abstract
Background/Objectives: Human glioblastoma (GBM) is the most aggressive brain cancer in adults and a highly treatment-refractory malignancy. The overall prognosis for the GBM is extremely poor, with a median survival of 12–14 months after initial diagnosis. Many GBM patients initially respond to [...] Read more.
Background/Objectives: Human glioblastoma (GBM) is the most aggressive brain cancer in adults and a highly treatment-refractory malignancy. The overall prognosis for the GBM is extremely poor, with a median survival of 12–14 months after initial diagnosis. Many GBM patients initially respond to the DNA alkylating agent temozolomide (TMZ), but patients often become therapy-resistant, and tumors recur. We previously reported that treatment with PFI-3, which is a small molecule inhibitor of the bromodomain of the BRG1 subunit of the SW1/SNF chromatin remodeling complex, enhanced the sensitivity of GBM cells to TMZ in vitro and in vivo GBM animal models. Our general objective was to perform an SAR study of new diphenyl PFI-3 analogs. Methods: New structural analogs of PFI-3 were developed, synthesized, and tested for their ability to enhance TMZ-induced GBM cell death by ELISA. Results: Following on the enhanced activity of compounds 2a and 2b, new diphenyl PFI-3 analogs with specific structural adjustments were made to better understand the structural requirements to optimize function. Additionally, several new structurally different candidates (e.g., 4a, 4b, and 5) showed much better efficacy in sensitizing GBM cells to TMZ-induced GBM cell death. Conclusions: Four series of PFI-3 analogs (2, 3, 4, and 5) were designed, synthesized, and tested for the ability to sensitize GBM cells to TMZ-induced cell death. Series 2 optimized the A-ring and R-isomer chirality. Series 3 used a 5-membered linker with weak activity. Series 4’s di-phenyl urea compounds showed better bromodomain inhibition. Series 5’s methoxyphenyl-B-ring analogs were exceptionally strong inhibitors. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

23 pages, 16798 KiB  
Article
Huafengdan Inhibits Glioblastoma Cell Growth and Mobility by Acting on PLAU and CAV1 Targets
by Dengxiao Lin, Wenfeng Yu, Jia Yu, Sha Cheng, Yu Song, Xiaoqing Wan, Yingjiang Xu, Heng Luo and Baofei Sun
Pharmaceuticals 2025, 18(3), 428; https://doi.org/10.3390/ph18030428 - 18 Mar 2025
Cited by 1 | Viewed by 612 | Correction
Abstract
Background: Glioblastoma (GBM) is considered a clinically refractory malignant tumor due to its high recurrence and malignancy, invasiveness, and poor prognosis. The ethnomedicine Huafengdan (HFD) is prepared using several Chinese herbs by a complex fermentation process that has a long history. Previous [...] Read more.
Background: Glioblastoma (GBM) is considered a clinically refractory malignant tumor due to its high recurrence and malignancy, invasiveness, and poor prognosis. The ethnomedicine Huafengdan (HFD) is prepared using several Chinese herbs by a complex fermentation process that has a long history. Previous studies have reported the inhibitory effect of HFD on GBM both in vitro and in vivo; however, its mechanism of action is unclear. Methods: The inhibitory effects of HFD on the growth, migration, and invasion of GBM cells were determined using the MTT assay, EdU assay, Transwell assay, flow cytometry, and Western blotting. A subcutaneous graft tumor model of nude BALB/c mice was established using U87 cells, and the in vivo activity and toxicity of HFD were evaluated using immunohistochemical staining and hematoxylin and eosin staining. Network pharmacology, bioinformatics, and transcriptomics were used to screen the targets and related signaling pathways of HFD in GBM and were validated using qPCR, CETSA, and Western blotting. Results: HFD inhibited the proliferation, invasion, and migration of GBM cells and induced S-phase block and apoptosis in GBM cells. It inhibited the in vivo growth of GBM cells without obvious toxicity. Mechanistic studies showed that the inhibition of GBM cell growth, migration, and invasion by HFD involved the key targets PLAU and CAV1. Its associated signaling pathways were the PI3K/Akt signaling pathway and cell cycle signaling pathway. Conclusions: Our findings confirm the novel function of HFD in inhibiting GBM cell growth in vitro and in vivo and highlight its potential in treating GBM. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

26 pages, 386 KiB  
Review
Expanding the Scope of Interventional Oncology: Locoregional Therapies in Extrahepatic Malignancies
by Gavin Wu, Cindy Chen, Jin Chang, Farbod Fazlollahi and Mina S. Makary
Cancers 2025, 17(5), 726; https://doi.org/10.3390/cancers17050726 - 21 Feb 2025
Viewed by 1216
Abstract
Background/Objectives: Locoregional therapies (LRTs), including transarterial embolization (TAE), transarterial chemoembolization (TACE), and transarterial radioembolization (TARE), have become integral in the management of hepatocellular carcinoma (HCC) in recent decades and continue to shape evolving treatment strategies. While their role in liver tumor management is [...] Read more.
Background/Objectives: Locoregional therapies (LRTs), including transarterial embolization (TAE), transarterial chemoembolization (TACE), and transarterial radioembolization (TARE), have become integral in the management of hepatocellular carcinoma (HCC) in recent decades and continue to shape evolving treatment strategies. While their role in liver tumor management is well established, their potential for treating extrahepatic malignancies is gaining increasing attention. Notably, growing research has highlighted the promising applications of TAE, TACE, and TARE in extrahepatic cancers such as glioblastoma (GBM), soft tissue sarcomas (STSs), prostate cancer (PCa), pancreatic cancer, and renal cell carcinoma (RCC). This review aims to explore these novel applications, providing a comprehensive summary of the current literature, examining clinical outcomes, and discussing future directions for integrating these techniques into broader oncologic treatment strategies. Methods: A systematic literature review was conducted focusing on LRTs such as TAE, TACE, and TARE in extrahepatic malignancies. Studies published between May 1998 and December 2024 were included, emphasizing outcomes in GBM, STS, PCa, pancreatic cancer, and RCC. Data extraction prioritized clinical outcomes, safety profiles, and procedural efficacy. Results: LRTs demonstrated significant potential in managing extrahepatic malignancies, with TAE, TACE, and TARE showing promising results in palliative management and tumor control. Across studies, these therapies exhibited varying degrees of success in improving progression-free survival and overall survival, with minimal systemic toxicity. Conclusions: The expanding application of LRTs in extrahepatic malignancies highlights their transformative potential in interventional oncology. By offering targeted, minimally invasive treatment options, these modalities bridge critical gaps in managing tumors refractory to conventional therapies. Future research should focus on standardizing protocols, optimizing patient selection, and exploring combination therapies to maximize their clinical efficacy. Full article
(This article belongs to the Special Issue Novel Approaches and Advances in Interventional Oncology)
12 pages, 958 KiB  
Case Report
Marine-Derived Therapeutics for the Management of Glioblastoma: A Case Series and Comprehensive Review of the Literature
by Nishika Karbhari and Simon Khagi
Onco 2024, 4(4), 369-380; https://doi.org/10.3390/onco4040026 - 4 Nov 2024
Cited by 1 | Viewed by 1936
Abstract
Introduction: Glioblastoma is a fatal intracranial neoplasm that is refractory to treatment, with inevitable disease recurrence and progression to death. Marine-derived compounds, including those found in nutraceutical products, may provide therapeutic benefit in the setting of glioblastoma. We present two patient cases whose [...] Read more.
Introduction: Glioblastoma is a fatal intracranial neoplasm that is refractory to treatment, with inevitable disease recurrence and progression to death. Marine-derived compounds, including those found in nutraceutical products, may provide therapeutic benefit in the setting of glioblastoma. We present two patient cases whose courses demonstrate a compelling role for marine-derived products in the management of glioblastoma. Cases: Case 1 describes a patient with MGMT promoter unmethylated glioblastoma who went on to complete standard of care chemoradiation along with concurrent use of a majority sea cucumber (MSC) blend known as SeaCare® (SeaCare, Torrington, CT, USA). Her survival of over 2 years significantly exceeds the recognized median survival time of glioblastoma. Case 2 describes a patient with a complicated course who experienced dramatic improvement after the initiation of the MSC blend, with an exceptional survival time of over 4 years post-diagnosis. Discussion: The mechanisms of marine-derived products that underlie these dramatic clinical effects are likely multifaceted but may hinge on the modification of the tumor immune microenvironment and suppression of tumorigenic effects. Specifically, the change in tumor-associated macrophages (TAMs) within the tumor microenvironment is central to this complex interplay. Conclusions: Ultimately, the use of marine products in the treatment of glioblastoma may present a novel and promising therapeutic strategy that warrants further investigation. Full article
Show Figures

Figure 1

9 pages, 681 KiB  
Review
State of the Art on CAR T-Cell Therapies for Onco-Haematological Disorders and Other Conditions
by Jose Alejandro Madrigal and José C. Crispín
Onco 2024, 4(3), 232-240; https://doi.org/10.3390/onco4030017 - 8 Sep 2024
Cited by 2 | Viewed by 2190
Abstract
The use of chimeric antigen receptors (CAR T-cells) for the treatment of patients with malignant haematological diseases has become a well-established application for conditions such as refractory or relapsed B-cell acute lymphoblastic leukaemia (B-ALL), B-cell lymphomas (BCL), and multiple myeloma (MM). Nearly 35,000 [...] Read more.
The use of chimeric antigen receptors (CAR T-cells) for the treatment of patients with malignant haematological diseases has become a well-established application for conditions such as refractory or relapsed B-cell acute lymphoblastic leukaemia (B-ALL), B-cell lymphomas (BCL), and multiple myeloma (MM). Nearly 35,000 patients have received autologous CAR T-cells for the treatment of these conditions only in the USA. Since their approval by the Food and Drug Administration (FDA) in 2017, over 1200 clinical trials have been initiated globally and there are at least 10 different CAR T-cells with approval by different regulatory agencies around the globe. In the USA, the FDA has approved six commercial CAR T-cells that are widely distributed worldwide. At the time of writing, several clinical trials have been performed in patients with solid tumours such as glioblastoma, renal and pancreatic cancer, as well as in patients with autoimmune conditions such as systemic lupus erythematosus (SLE), idiopathic inflammatory myositis (IIM), and systemic sclerosis (SS). There are also several studies showing the potential benefit of CAR T-cells for other non-malignant diseases such as asthma and even fungal infections. In this review, without pretending to cover all current areas of treatments with CAR T-cells, we offer a brief summary of some of the most relevant aspects of the use of CAR T-cells for some of these conditions. Full article
Show Figures

Figure 1

17 pages, 770 KiB  
Review
Phagocytosis Checkpoints in Glioblastoma: CD47 and Beyond
by Amber Afzal, Zobia Afzal, Sophia Bizink, Amanda Davis, Sara Makahleh, Yara Mohamed and Salvatore J. Coniglio
Curr. Issues Mol. Biol. 2024, 46(8), 7795-7811; https://doi.org/10.3390/cimb46080462 - 23 Jul 2024
Cited by 2 | Viewed by 3493
Abstract
Glioblastoma multiforme (GBM) is one of the deadliest human cancers with very limited treatment options available. The malignant behavior of GBM is manifested in a tumor which is highly invasive, resistant to standard cytotoxic chemotherapy, and strongly immunosuppressive. Immune checkpoint inhibitors have recently [...] Read more.
Glioblastoma multiforme (GBM) is one of the deadliest human cancers with very limited treatment options available. The malignant behavior of GBM is manifested in a tumor which is highly invasive, resistant to standard cytotoxic chemotherapy, and strongly immunosuppressive. Immune checkpoint inhibitors have recently been introduced in the clinic and have yielded promising results in certain cancers. GBM, however, is largely refractory to these treatments. The immune checkpoint CD47 has recently gained attention as a potential target for intervention as it conveys a “don’t eat me” signal to tumor-associated macrophages (TAMs) via the inhibitory SIRP alpha protein. In preclinical models, the administration of anti-CD47 monoclonal antibodies has shown impressive results with GBM and other tumor models. Several well-characterized oncogenic pathways have recently been shown to regulate CD47 expression in GBM cells and glioma stem cells (GSCs) including Epidermal Growth Factor Receptor (EGFR) beta catenin. Other macrophage pathways involved in regulating phagocytosis including TREM2 and glycan binding proteins are discussed as well. Finally, chimeric antigen receptor macrophages (CAR-Ms) could be leveraged for greatly enhancing the phagocytosis of GBM and repolarization of the microenvironment in general. Here, we comprehensively review the mechanisms that regulate the macrophage phagocytosis of GBM cells. Full article
(This article belongs to the Special Issue Future Challenges of Targeted Therapy of Cancers: 2nd Edition)
Show Figures

Figure 1

18 pages, 4233 KiB  
Article
IMPDH Inhibition Decreases TERT Expression and Synergizes the Cytotoxic Effect of Chemotherapeutic Agents in Glioblastoma Cells
by Xiaoqin Liu, Junying Wang, Laura J. Wu, Britni Trinh and Robert Y. L. Tsai
Int. J. Mol. Sci. 2024, 25(11), 5992; https://doi.org/10.3390/ijms25115992 - 30 May 2024
Cited by 5 | Viewed by 1923
Abstract
IMP dehydrogenase (IMPDH) inhibition has emerged as a new target therapy for glioblastoma multiforme (GBM), which remains one of the most refractory tumors to date. TCGA analyses revealed distinct expression profiles of IMPDH isoenzymes in various subtypes of GBM and low-grade glioma (LGG). [...] Read more.
IMP dehydrogenase (IMPDH) inhibition has emerged as a new target therapy for glioblastoma multiforme (GBM), which remains one of the most refractory tumors to date. TCGA analyses revealed distinct expression profiles of IMPDH isoenzymes in various subtypes of GBM and low-grade glioma (LGG). To dissect the mechanism(s) underlying the anti-tumor effect of IMPDH inhibition in adult GBM, we investigated how mycophenolic acid (MPA, an IMPDH inhibitor) treatment affected key oncogenic drivers in glioblastoma cells. Our results showed that MPA decreased the expression of telomerase reverse transcriptase (TERT) in both U87 and U251 cells, and the expression of O6-methylguanine-DNA methyltransferase (MGMT) in U251 cells. In support, MPA treatment reduced the amount of telomere repeats in U87 and U251 cells. TERT downregulation by MPA was associated with a significant decrease in c-Myc (a TERT transcription activator) in U87 but not U251 cells, and a dose-dependent increase in p53 and CCCTC-binding factor (CTCF) (TERT repressors) in both U87 and U251 cells. In U251 cells, MPA displayed strong cytotoxic synergy with BCNU and moderate synergy with irinotecan, oxaliplatin, paclitaxel, or temozolomide (TMZ). In U87 cells, MPA displayed strong cytotoxic synergy with all except TMZ, acting primarily through the apoptotic pathway. Our work expands the mechanistic potential of IMPDH inhibition to TERT/telomere regulation and reveals a synthetic lethality between MPA and anti-GBM drugs. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

21 pages, 1483 KiB  
Review
The Role of Mesenchymal Reprogramming in Malignant Clonal Evolution and Intra-Tumoral Heterogeneity in Glioblastoma
by Qiong Wu, Anders E. Berglund, Robert J. Macaulay and Arnold B. Etame
Cells 2024, 13(11), 942; https://doi.org/10.3390/cells13110942 - 30 May 2024
Cited by 5 | Viewed by 1883
Abstract
Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated [...] Read more.
Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated with significantly increased long-term survival compared to the highly resistant mesenchymal (MES) subtype. Furthermore, it is universally recognized that a small subset of GBM cells known as GBM stem cells (GSCs) serve as reservoirs for tumor recurrence and progression. The clonal evolution of GSC molecular subtypes in response to therapy drives intra-tumoral heterogeneity and remains a critical determinant of GBM outcomes. In particular, the intra-tumoral MES reprogramming of GSCs using current GBM therapies has emerged as a leading hypothesis for therapeutic refractoriness. Preventing the intra-tumoral divergent evolution of GBM toward the MES subtype via new treatments would dramatically improve long-term survival for GBM patients and have a significant impact on GBM outcomes. In this review, we examine the challenges of the role of MES reprogramming in the malignant clonal evolution of glioblastoma and provide future perspectives for addressing the unmet therapeutic need to overcome resistance in GBM. Full article
(This article belongs to the Special Issue Cell Death Mechanisms and Therapeutic Opportunities in Glioblastoma)
Show Figures

Figure 1

22 pages, 3802 KiB  
Review
The Role of Bacteria in Central Nervous System Tumors: Opportunities and Challenges
by Rui Zhang, Xueying Li and Si Zhang
Microorganisms 2024, 12(6), 1053; https://doi.org/10.3390/microorganisms12061053 - 23 May 2024
Cited by 3 | Viewed by 3005
Abstract
Tumors of the central nervous system (CNS) are severe and refractory diseases with poor prognosis, especially for patients with malignant glioblastoma and brain metastases. Currently, numerous studies have explored the potential role of bacteria and intestinal flora in tumor development and treatment. Bacteria [...] Read more.
Tumors of the central nervous system (CNS) are severe and refractory diseases with poor prognosis, especially for patients with malignant glioblastoma and brain metastases. Currently, numerous studies have explored the potential role of bacteria and intestinal flora in tumor development and treatment. Bacteria can penetrate the blood–brain barrier (BBB), targeting the hypoxic microenvironment at the core of tumors, thereby eliminating tumors and activating both the innate and adaptive immune responses, rendering them promising therapeutic agents for CNS tumors. In addition, engineered bacteria and derivatives, such as bacterial membrane proteins and bacterial spores, can also be used as good candidate carriers for targeted drug delivery. Moreover, the intestinal flora can regulate CNS tumor metabolism and influence the immune microenvironment through the “gut–brain axis”. Therefore, bacterial anti-tumor therapy, engineered bacterial targeted drug delivery, and intervention of the intestinal flora provide therapeutic modalities for the treatment of CNS tumors. In this paper, we performed a comprehensive review of the mechanisms and therapeutic practices of bacterial therapy for CNS tumors and discussed potential future research directions in this field. Full article
(This article belongs to the Special Issue Advances in Medical Microbiology)
Show Figures

Graphical abstract

15 pages, 2288 KiB  
Article
Therapeutic Potential of Cannabinoid Profiles Identified in Cannabis L. Crops in Peru
by Pedro Wong-Salgado, Fabiano Soares, Jeel Moya-Salazar, José F. Ramírez-Méndez, Marcia M. Moya-Salazar, Alfonso Apesteguía and Americo Castro
Biomedicines 2024, 12(2), 306; https://doi.org/10.3390/biomedicines12020306 - 29 Jan 2024
Cited by 2 | Viewed by 4207
Abstract
Cannabis is a plant that is cultivated worldwide, and its use is internationally regulated, but some countries have been regulating its medicinal, social, and industrial uses. This plant must have arrived in Peru during the Spanish conquest and remains widely cultivated illicitly or [...] Read more.
Cannabis is a plant that is cultivated worldwide, and its use is internationally regulated, but some countries have been regulating its medicinal, social, and industrial uses. This plant must have arrived in Peru during the Spanish conquest and remains widely cultivated illicitly or informally to this day. However, new regulations are currently being proposed to allow its legal commercialization for medicinal purposes. Cannabis contains specific metabolites known as cannabinoids, some of which have clinically demonstrated therapeutic effects. It is now possible to quantitatively measure the presence of these cannabinoids in dried inflorescences, thus allowing for description of the chemical profile or “chemotype” of cannabinoids in each sample. This study analyzed the chemotypes of eight samples of dried inflorescences from cannabis cultivars in four different regions of Peru, and based on the significant variation in the cannabinoid profiles, we suggest their therapeutic potential. The most important medical areas in which they could be used include the following: they can help manage chronic pain, they have antiemetic, anti-inflammatory, and antipruritic properties, are beneficial in treating duodenal ulcers, can be used in bronchodilators, in muscle relaxants, and in treating refractory epilepsy, have anxiolytic properties, reduce sebum, are effective on Methicillin-resistant Staphylococcus aureus, are proapoptotic in breast cancer, can be used to treat addiction and psychosis, and are effective on MRSA, in controlling psoriasis, and in treating glioblastoma, according to the properties of their concentrations of cannabidiol, cannabigerol, and Δ9-tetrahydrocannabinol, as reviewed in the literature. On the other hand, having obtained concentrations of THC, we were able to suggest the psychotropic capacity of said samples, one of which even fits within the legal category of “non-psychoactive cannabis” according to Peruvian regulations. Full article
(This article belongs to the Special Issue Therapeutic Potential for Cannabis and Cannabinoids 2.0)
Show Figures

Figure 1

12 pages, 1159 KiB  
Review
Unlocking Glioblastoma Secrets: Natural Killer Cell Therapy against Cancer Stem Cells
by Yuanning Du, Karen E. Pollok and Jia Shen
Cancers 2023, 15(24), 5836; https://doi.org/10.3390/cancers15245836 - 14 Dec 2023
Cited by 9 | Viewed by 3110
Abstract
Glioblastoma (GBM) represents a paramount challenge as the most formidable primary brain tumor characterized by its rapid growth, aggressive invasiveness, and remarkable heterogeneity, collectively impeding effective therapeutic interventions. The cancer stem cells within GBM, GBM stem cells (GSCs), hold pivotal significance in fueling [...] Read more.
Glioblastoma (GBM) represents a paramount challenge as the most formidable primary brain tumor characterized by its rapid growth, aggressive invasiveness, and remarkable heterogeneity, collectively impeding effective therapeutic interventions. The cancer stem cells within GBM, GBM stem cells (GSCs), hold pivotal significance in fueling tumor advancement, therapeutic refractoriness, and relapse. Given their unique attributes encompassing self-renewal, multipotent differentiation potential, and intricate interplay with the tumor microenvironment, targeting GSCs emerges as a critical strategy for innovative GBM treatments. Natural killer (NK) cells, innate immune effectors recognized for their capacity to selectively detect and eliminate malignancies without the need for prior sensitization, offer substantial therapeutic potential. Harnessing the inherent capabilities of NK cells can not only directly engage tumor cells but also augment broader immune responses. Encouraging outcomes from clinical investigations underscore NK cells as a potentially effective modality for cancer therapy. Consequently, NK cell-based approaches hold promise for effectively targeting GSCs, thereby presenting an avenue to enhance treatment outcomes for GBM patients. This review outlines GBM’s intricate landscape, therapeutic challenges, GSC-related dynamics, and elucidates the potential of NK cell as an immunotherapeutic strategy directed towards GSCs. Full article
(This article belongs to the Special Issue Brain Cancer Stem Cells in Children and Adults)
Show Figures

Figure 1

17 pages, 3989 KiB  
Article
Fibronectin Type III Domain Containing 3B as a Potential Prognostic and Therapeutic Biomarker for Glioblastoma
by Hyukjun Kwon, Minji Yun, Taek-Hyun Kwon, Minji Bang, Jungsul Lee, Yeo Song Lee, Hae Young Ko and Kyuha Chong
Biomedicines 2023, 11(12), 3168; https://doi.org/10.3390/biomedicines11123168 - 28 Nov 2023
Cited by 5 | Viewed by 2367
Abstract
Glioblastoma (GBM) is a representative malignant brain tumor characterized by a dismal prognosis, with survival rates of less than 2 years and high recurrence rates. Despite surgical resection and several alternative treatments, GBM remains a refractory disease due to its aggressive invasiveness and [...] Read more.
Glioblastoma (GBM) is a representative malignant brain tumor characterized by a dismal prognosis, with survival rates of less than 2 years and high recurrence rates. Despite surgical resection and several alternative treatments, GBM remains a refractory disease due to its aggressive invasiveness and resistance to anticancer therapy. In this report, we explore the role of fibronectin type III domain containing 3B (FNDC3B) and its potential as a prognostic and therapeutic biomarker in GBM. GBM exhibited a significantly higher cancer-to-normal ratio compared to other organs, and patients with high FNDC3B expression had a poor prognosis (p < 0.01). In vitro studies revealed that silencing FNDC3B significantly reduced the expression of Survivin, an apoptosis inhibitor, and also reduced cell migration, invasion, extracellular matrix adhesion ability, and stem cell properties in GBM cells. Furthermore, we identified that FNDC3B regulates PTEN/PI3K/Akt signaling in GBM cells using MetaCore integrated pathway bioinformatics analysis and a proteome profiler phospho-kinase array with sequential western blot analysis. Collectively, our findings suggest FNDC3B as a potential biomarker for predicting GBM patient survival and for the development of treatment strategies for GBM. Full article
Show Figures

Graphical abstract

20 pages, 2027 KiB  
Article
A Multicenter Randomized Bioequivalence Study of a Novel Ready-to-Use Temozolomide Oral Suspension vs. Temozolomide Capsules
by François Ducray, Carole Ramirez, Marie Robert, Maxime Fontanilles, Charlotte Bronnimann, Olivier Chinot, Florian Estrade, Xavier Durando, Stéphanie Cartalat, Jeremy Bastid, Hugues Bienayme and Caroline Lemarchand
Pharmaceutics 2023, 15(12), 2664; https://doi.org/10.3390/pharmaceutics15122664 - 24 Nov 2023
Viewed by 2821
Abstract
Background: Temozolomide (TMZ) oral suspension (Ped-TMZ, KIZFIZO®) is being developed for the treatment of relapsed or refractory neuroblastoma, a rare cancer affecting infants and young children. The study assessed the safety and the bioequivalence of this novel pediatric formulation with existing [...] Read more.
Background: Temozolomide (TMZ) oral suspension (Ped-TMZ, KIZFIZO®) is being developed for the treatment of relapsed or refractory neuroblastoma, a rare cancer affecting infants and young children. The study assessed the safety and the bioequivalence of this novel pediatric formulation with existing TMZ oral capsules. Methods: In vitro dissolution profiles and the bioequivalence were evaluated following the European Medicines Agency “Guidelines on the investigation of Bioequivalence”. The phase I, multicenter, randomized, open-label, crossover, single-dose bioequivalence study enrolled 36 adult patients with glioblastoma multiforme or lower-grade glioma. Each patient received 200 mg/m2 Ped-TMZ suspension and TMZ capsules (Temodal®) on 2 consecutive days, with the order being randomly assigned. Fourteen blood samples were collected up to 10 h post-dosing. Bioequivalence was assessed by comparing the 90% confidence interval for the ratio of the geometric means of maximum TMZ plasma concentration (Cmax) and the area under the curve (AUCt). Other endpoints included further pharmacokinetic parameters and safety. Results: Both formulations exhibited a fast in vitro dissolution profile with more than 85% of TMZ dissolved within 15 min. For the bioequivalence study, thirty patients completed the trial as per the protocol. The ratio of Ped-TMZ/TMZ capsule geometric means (90% CI) for AUCt and Cmax were 97.18% (95.05–99.35%) and 107.62% (98.07–118.09%), respectively, i.e., within the 80–125% bioequivalence limits. No buccal toxicity was associated with Ped-TMZ liquid formulation. Conclusions: This study showed that Ped-TMZ oral suspension and TMZ oral capsule treatment are immediate release and bioequivalent medicines. There were also no unexpected safety signals or local toxicity (funded by ORPHELIA Pharma; ClinicalTrials.gov number, NCT04467346). Full article
Show Figures

Figure 1

27 pages, 5890 KiB  
Review
The Clinical Validation of Modulated Electro-Hyperthermia (mEHT)
by Sun-Young Lee, Gergo Lorant, Laszlo Grand and Attila Marcell Szasz
Cancers 2023, 15(18), 4569; https://doi.org/10.3390/cancers15184569 - 15 Sep 2023
Cited by 9 | Viewed by 4008
Abstract
The mEHT method uses tissues’ thermal and bioelectromagnetic heterogeneity for the selective mechanisms. The success of the therapy for advanced, relapsed, and metastatic aggressive tumors can only be demonstrated by measuring survival time and quality of life (QoL). The complication is that mEHT-treated [...] Read more.
The mEHT method uses tissues’ thermal and bioelectromagnetic heterogeneity for the selective mechanisms. The success of the therapy for advanced, relapsed, and metastatic aggressive tumors can only be demonstrated by measuring survival time and quality of life (QoL). The complication is that mEHT-treated patients cannot be curatively treated any longer with “gold standards”, where the permanent progression of the disease, the refractory, relapsing situation, the organ failure, the worsening of blood counts, etc., block them. Collecting a cohort of these patients is frequently impossible. Only an intent-to-treat (ITT) patient group was available. Due to the above limitations, many studies have single-arm data collection. The Phase III trial of advanced cervix tumors subgrouping of HIV-negative and -positive patients showed the stable efficacy of mEHT in all patients’ subgroups. The single-arm represents lower-level evidence, which can be improved by comparing the survival data of various studies from different institutes. The Kaplan–Meier probability comparison had no significant differences, so pooled data were compared to other methods. Following this approach, we demonstrate the feasibility and superiority of mEHT in the cases of glioblastoma multiform, pancreas carcinomas, lung tumors, and colorectal tumors. Full article
(This article belongs to the Special Issue Advanced Research in Oncology in 2023)
Show Figures

Graphical abstract

Back to TopTop