1
|
Chithra P, Bhatia D, Solanki R. Advanced nanomicelles for targeted glioblastoma multiforme therapy. BIOMATERIALS ADVANCES 2025; 170:214221. [PMID: 39922136 DOI: 10.1016/j.bioadv.2025.214221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and malignant primary brain tumor, classified as grade IV by the WHO. Despite standard treatments like surgical resection, radiotherapy and chemotherapy (i.e. temozolomide), GBM's prognosis remains poor due to its heterogeneity, recurrence and the impermeability of the blood-brain barrier (BBB). The exact cause of GBM is unclear with potential factors including genetic predisposition and ionizing radiation. Innovative approaches such as nanomicelles-nanoscale, self-assembled structures made from lipids and amphiphilic polymers show promise for GBM therapy. These nanocarriers enhance drug solubility and stability, enabling targeted delivery of therapeutic agents across the BBB. This review explores the synthesis strategies, characterization and applications of nanomicelles in GBM treatment. Nanomicelles improve the delivery of both hydrophobic and hydrophilic drugs and provide non-invasive delivery options. By offering site-specific targeting, biocompatibility, and stability, nanomicelles can potentially overcome the limitations of current GBM therapies. This review highlights recent advancements in the use of nanomicelles for delivering therapeutic agents and nucleic acids addressing the critical need for advanced treatments to improve GBM patient outcomes.
Collapse
Affiliation(s)
- P Chithra
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
2
|
Rana M, Liou KC, Thakur A, Nepali K, Liou JP. Advancing glioblastoma therapy: Learning from the past and innovations for the future. Cancer Lett 2025; 617:217601. [PMID: 40037502 DOI: 10.1016/j.canlet.2025.217601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Marred by a median survival of only around 12-15 months coupled with poor prognosis and effective therapeutic deprived drug armory, treatment/management of glioblastoma has proved to be a daunting task. Surgical resection, flanked by radiotherapy and chemotherapy with temozolomide, stands as the standard of care; however, this trimodal therapy often manifests limited efficacy due to the heterogeneous and highly infiltrative nature of GBM cells. In addition, the existence of the blood-brain barrier, tumor microenvironment, and the immunosuppressive nature of GBM, along with the encountered resistance of GBM cells towards conventional therapy, also hinders the therapeutic applications of chemotherapeutics in GBM. This review presents key insights into the molecular pathology of GBM, including genetic mutations, signaling pathways, and tumor microenvironment characteristics. Recent innovations such as immunotherapy, oncolytic viral therapies, vaccines, nanotechnology, electric field, and cancer neuroscience, as well as their clinical progress, have been covered. In addition, this compilation also encompasses a discussion on the role of personalized medicine in tailoring treatments based on individual tumor profiles, an approach that is gradually shifting the paradigm in GBM management. Endowed with the learnings imbibed from past failures coupled with the zeal to embrace novel/multidisciplinary approaches, researchers appear to be on the right track to pinpoint more effective and durable solutions in the context of GBM treatment.
Collapse
Affiliation(s)
- Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Ke-Chi Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
3
|
Ceresoli GL, Gianoncelli L. Tumor Treating Fields (TTFields) Therapy in Unresectable Pleural Mesothelioma: Overview of Efficacy, Safety, and Future Outlook. Curr Treat Options Oncol 2025:10.1007/s11864-025-01320-w. [PMID: 40266436 DOI: 10.1007/s11864-025-01320-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
OPINION STATEMENT Pleural mesothelioma is an incurable cancer with unmet diagnostic and therapeutic needs. Due to its pattern of local spread, few patients are candidates for multimodality treatment and thus most patients only receive systemic therapy. Chemotherapy (pemetrexed plus platinum) was standard of care until the recent addition of immunotherapy (nivolumab plus ipilimumab, or pembrolizumab plus chemotherapy) as further first-line option. Physicians treating pleural mesothelioma should be aware of another option with Tumor Treating Fields (TTFields) therapy, a locoregionally-applied therapy utilizing electric fields generated by a portable medical device, and delivered to the tumor by skin-placed arrays. TTFields therapy delivered to the thorax using the NovoTTF- 100L device concomitant with pemetrexed and platinum agent is approved for unresectable pleural mesothelioma in the US, and received Conformité Européenne certification in Europe, based on results from the phase 2 STELLAR study (EF- 23; NCT02397928), where TTFields-related toxicity was limited to mild-to-moderate reversible skin reactions. Overall survival in the STELLAR study with TTFields therapy was 18.2 months, with further post-hoc analysis showing extended survival in patients with epithelioid histology. Within the evolving landscape of systemic treatments, TTFields therapy represents a novel and clinically versatile therapeutic option in the battle against pleural mesothelioma without introducing additional toxicities other than mild-to-moderate skin irritation. While promising, additional research is needed to optimize clinical application of TTFields therapy in patients with pleural mesothelioma, such as identifying the molecular determinants of therapy efficacy, and further investigation into the safe and effective delivery of TTFields therapy together with systemic agents, including immunotherapies.
Collapse
Affiliation(s)
- Giovanni Luca Ceresoli
- Medical Oncology Unit, Cliniche Humanitas Gavazzeni, Via Mauro Gavazzeni, 21, Bergamo, Italy.
| | - Letizia Gianoncelli
- Medical Oncology Unit, ASST Santi Paolo E Carlo, Ospedale San Paolo, Via Antonio Di Rudinì, 8, 20124, Milan, Italy
| |
Collapse
|
4
|
Rodella G, Ma Z, Ucakar B, Joudiou N, Préat V, Gallez B, Malfanti A. Repurposing Chemotherapeutics in a Hyaluronic Acid-conjugate Combination Treatment Approach for the Local Immunomodulation of the Glioblastoma Microenvironment. Int J Pharm 2025; 676:125612. [PMID: 40252866 DOI: 10.1016/j.ijpharm.2025.125612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
The immunosuppressive tumor immune microenvironment (TIME) renders glioblastoma (GBM) refractory to current chemo-immunotherapeutics. We sought to explore a novel approach for local GBM-associated TIME immunomodulation based on a synergistic combination of the repurposed chemotherapeutic drugs doxorubicin (DOX), which acts to induce immunogenic cell death (ICD) and gemcitabine (GEM), which depletes immunosuppressive myeloid-derived suppressor cells (MDSCs). We conjugated DOX and GEM to hyaluronic acid (HA) to improve efficacy, given this polymer's ability to target CD44 which are overexpressed on cancer cells. The HA-DOX and HA-GEM polymer-drug conjugates provided synergistic cytotoxic effects and maintained ICD-related properties in GBM cells compared to a combination of free drugs. HA-DOX and HA-GEM also reverted the immunosuppressive GBM-associated TIME in orthotopic GL261 tumor-bearing mice by selectively depleting MDSCs and reprogramming M2-like macrophages towards a pro-inflammatory M1-like state, resulting in controlled tumor growth. Local HA-DOX and HA-GEM delivery also increased median survival and controlled tumor growth in an immune refractory SB28-GBM orthotopic mouse GBM model. These findings highlight the potential of repurposing clinically applicable chemotherapeutics in the context of polymer-drug combination treatments for novel immunomodulation strategies in unresectable GBM, which may open new avenues for developing innovative therapies.
Collapse
Affiliation(s)
- Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Zhanjun Ma
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Ucakar
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Nicolas Joudiou
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; Departement of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo, 5, 35131 Padova, Italy.
| |
Collapse
|
5
|
Feng S, Liu H, Yun C, Zhu W, Pan Y. Application of EGFR-TKIs in brain tumors, a breakthrough in future? J Transl Med 2025; 23:449. [PMID: 40241139 PMCID: PMC12004797 DOI: 10.1186/s12967-025-06448-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Brain tumors, both primary and secondary, represent a significant clinical challenge due to their high mortality and limited treatment options. Primary brain tumors, such as gliomas and meningiomas, and brain metastases from cancers such as non-small cell lung cancer and breast cancer require innovative therapeutic strategies. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR -TKIs) have emerged as a promising treatment option, particularly for tumors harboring EGFR mutations. This review examines the use of EGFR-TKIs in brain tumors, highlighting both laboratory and clinical research findings. In primary brain tumors and brain metastases, EGFR-TKIs have shown potential in controlling tumor growth and improving patient outcomes. Advanced applications, such as nano-formulated EGFR-TKIs and combination therapies with other pathway inhibitors, are being investigated to improve efficacy and overcome resistance. Challenges such as treatment-related events, resistance mechanisms and blood-brain barrier penetration remain significant hurdles. Addressing tumor heterogeneity through personalized medicine approaches is critical to optimizing EGFR-TKI therapies. This review highlights the need for continued research to refine these therapies and improve survival for patients with brain tumors.
Collapse
Affiliation(s)
- Shiying Feng
- Central Clinical Medical School, Baotou Medical College, Baotou, Inner Mongolia, 014040, China
- Department of Oncology, Inner Mongolia Baotou City Central Hospital, Baotou, Inner Mongolia, 014040, China
| | - Huiqin Liu
- Department of Gynecology & Obstetrics, Inner Mongolia Baotou City Central Hospital, Baotou, Inner Mongolia, 014040, China
| | - Cuilan Yun
- Department of Gynecology & Obstetrics, Inner Mongolia Baotou City Central Hospital, Baotou, Inner Mongolia, 014040, China
| | - Wei Zhu
- Department of Oncology, Inner Mongolia Baotou City Central Hospital, Baotou, Inner Mongolia, 014040, China.
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
6
|
Liu RN, Huang JH, Qi X, Pan Y, Wu E, Nizamutdinov D. Tumor Treating Fields and Combination Therapy in Management of Brain Oncology. Cancers (Basel) 2025; 17:1211. [PMID: 40227773 PMCID: PMC11987984 DOI: 10.3390/cancers17071211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
Glioblastoma (GBM) remains a challenging cancer to treat with limited effective therapies. Standard treatments, including surgery, radiotherapy, chemotherapy, targeted therapy, and immunotherapy, offer marginal survival benefits but are often limited by side effects and drug resistance. Temozolomide is the most commonly used chemotherapy; however, resistance and lack of efficacy in recurrent GBM hinder its success. Tumor treating fields (TTFields), a novel non-invasive modality that utilizes alternating electric fields, have recently emerged as a promising treatment for GBM. TTFields work by disrupting the function of the mitotic spindle and inducing apoptosis in cancer cells. They can be especially effective when combined with other therapies. TTFields enhance drug delivery when paired with chemotherapy by increasing the permeability of the blood-brain barrier and cell membranes, leading to more effective tumor inhibition. Similarly, TTFields increase cancer cell sensitivity to radiation therapy and improve the efficacy of targeted therapies, such as sorafenib and immunotherapy, particularly in extra-cranial tumors. The Optune device, the primary medical device for TTFields' delivery, offers a convenient and versatile treatment option, allowing remote care and exhibiting fewer adverse effects. This review discusses the potential of TTFields as a valuable addition to GBM treatment, particularly in combination therapies, and highlights the device's clinical applications.
Collapse
Affiliation(s)
- Ruisi Nicole Liu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
| | - James H. Huang
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
| | - Xiaoming Qi
- Department of Neurology, Baylor Scott & White Health, Temple, TX 76508, USA
| | - Yizhong Pan
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
- Department of Neurosurgery, First Affiliated Hospital of Soochow University, Suzhou 215005, China
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX 76508, USA
| | - Damir Nizamutdinov
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
| |
Collapse
|
7
|
Matsuzaki H, Kai K, Komohara Y, Yano H, Pan C, Fujiwara Y, Yamada R, Iwauchi A, Fukasawa N, Tanaka T, Shimoda M, Watanabe H, Maruyama T, Takeo T, Mikami Y, Mukasa A. Abnormal Vessels Potentially Accelerate Glioblastoma Proliferation by Inducing the Protumor Activation of Macrophages. Cancer Sci 2025; 116:897-909. [PMID: 39921277 PMCID: PMC11967248 DOI: 10.1111/cas.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
Glioblastoma (GBM) involves disruptions in the blood-brain barrier (BBB) and alterations in the immune microenvironment, including the activation of glioma-associated macrophages (GAMs). Vascular endothelial growth factor inhibitors, commonly used in recurrent GBM treatment, can influence these processes. This study investigates the relationship between BBB disruption and GAM activation, focusing on plasmalemma vesicle-associated protein (PLVAP), a marker of BBB disruption, and α1-acid glycoprotein (AGP), an inflammatory protein implicated in tumor progression. PLVAP expression was analyzed by immunohistochemistry (IHC) in human GBM samples to determine correlations with tumor grade, proliferation, and GAM activation. Pre- and post-bevacizumab treatment GBM samples were compared to assess changes in BBB integrity and macrophage activity. AGP's role in GAM activation was studied through in vitro assays and glioma implantation in AGP knockout mice, with assessments of tumor growth and angiogenesis. Results showed elevated PLVAP expression in higher-grade gliomas, correlating with increased tumor proliferation and GAM activation, particularly around PLVAP-positive vessels. Bevacizumab treatment reduced PLVAP expression and macrophage activity. AGP localized to regions of BBB disruption, promoting macrophage-mediated tumor growth in vitro. AGP knockout mice demonstrated reduced angiogenesis and prolonged survival. Spatial analysis revealed increased expression of macrophage-inducing molecules near PLVAP-positive vessels. These findings suggest PLVAP as a marker of BBB disruption and glioma malignancy. AGP, associated with BBB leakage, contributes to GAM activation and tumor progression, highlighting its potential as a therapeutic target for GBM.
Collapse
Affiliation(s)
- Hiroaki Matsuzaki
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Neurosurgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Keitaro Kai
- Department of Neurosurgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Tumor Pathology, Graduate School of Health SciencesKumamoto UniversityKumamotoJapan
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Rin Yamada
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Diagnostic Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Ai Iwauchi
- Department of PathologyThe Jikei University School of MedicineTokyoJapan
| | - Nei Fukasawa
- Department of PathologyThe Jikei University School of MedicineTokyoJapan
| | - Toshihide Tanaka
- Department of NeurosurgeryThe Jikei University School of MedicineTokyoJapan
| | - Masayuki Shimoda
- Department of PathologyThe Jikei University School of MedicineTokyoJapan
| | - Hiroshi Watanabe
- Department of Clinical Pharmacy and Therapeutics, Graduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and DevelopmentKumamoto UniversityKumamotoJapan
| | - Yoshiki Mikami
- Department of Diagnostic Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Akitake Mukasa
- Department of Neurosurgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
8
|
Narang K, Kataria T, Bisht SS, Gupta D, Banerjee S, Mayank M, Shishak S, Kaliyaperumal V, Tamilselvan S, Kamaraj D, Abraham S. Contemporary Long-term Survival Outcomes and Prognostic Factors in Adult grade 4 Astrocytoma: An Institutional Analysis. Clin Oncol (R Coll Radiol) 2025; 40:103788. [PMID: 40048926 DOI: 10.1016/j.clon.2025.103788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 03/29/2025]
Abstract
AIMS Astrocytoma grade 4 without isocitrate dehydrogenase (IDH)-based characterisation has been called glioblastoma (GBM) in historical cohorts. There have been significant advancements in diagnostic radiology and pathology, and in the technical aspects of surgery, radiation therapy, and temozolomide (TMZ) used for treatment of this disease. We analysed the outcomes of 267 adult astrocytoma grade 4/GBM patients, consecutively treated between December 2010 and November 2018 using modern techniques at our institute. MATERIALS AND METHODS All patients underwent surgical resection, histopathology review, and O6-methylguanine-DNA methyltransferase (MGMT) methylation testing, volumetric modulated arc therapy (VMAT)-based radiation therapy using institute-specific target-delineation guidelines and image guidance, and TMZ according to Stupp protocol. Serial multiparametric magnetic resonance imaging-based follow-up ensured early detection of disease progression. Appropriate salvage therapy was determined based on clinicopathological attributes. Kaplan-Meier survival plots, log-rank test, and Cox regression analysis were performed on the prospectively recorded dataset to estimate survival and the factors affecting it. RESULTS At a median follow-up of 72 months, the median progression-free survival (PFS), 1-year PFS, and 2-year PFS were 10 months, 37.8%, and 17.5%, respectively. MGMT-methylation, a radiation dose ≥54 Gy, and ≥4 adjuvant TMZ cycles were associated with favourable PFS. Median overall survival (OS), 2-year OS and 5-year OS were 24 months, 48%, and 18%, respectively. MGMT-methylation and 1-year disease control were associated with favourable OS. Salvage treatment could be offered to 69.2% patients, with use of all the three treatment modalities in 12.4%. Salvage reirradiation could be used in 30.8% patients. Haematological toxicity ≥grade 2 was evident in 6% patients during concurrent radiation-TMZ phase and in 9% patients in adjuvant TMZ phase. Postradiation neurocognitive deficits were noted in 20.1% patients, with onset at a median duration of 10 months. CONCLUSION Modern diagnostic and therapeutic techniques affected a near-doubling of survival and acceptable late toxicity, as compared to historical data.
Collapse
Affiliation(s)
- K Narang
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India.
| | - T Kataria
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S S Bisht
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - D Gupta
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Banerjee
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - M Mayank
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Shishak
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - V Kaliyaperumal
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Tamilselvan
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - D Kamaraj
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Abraham
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| |
Collapse
|
9
|
Zhai Y, Li G, Pan C, Yu M, Hu H, Wang D, Shi Z, Jiang T, Zhang W. The development and potent antitumor efficacy of CD44/CD133 dual-targeting IL7Rα-armored CAR-T cells against glioblastoma. Cancer Lett 2025; 614:217541. [PMID: 39952598 DOI: 10.1016/j.canlet.2025.217541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Tumor heterogeneity and an immunosuppressive microenvironment pose significant challenges for immunotherapy against solid tumors, particularly glioblastoma multiforme (GBM). Recent studies have highlighted the crucial role of glioma stem cells (GSCs) in tumor recurrence and therapeutic resistance. In this context, we developed a tandem chimeric antigen receptor (CAR)-T cell targeting CD44 and CD133 (PROM1), containing a truncated IL-7 receptor alpha intracellular domain (Δ7R) between the CD28 costimulatory receptor and the CD3ζ signaling chain (Tanζ-T28-Δ7R). Our target identification and validation were carried out using GSCs, samples from GBM patients, and the corresponding sequencing data. The antitumor efficacy of CAR-T cells was evaluated in patient-derived GSCs, intracranial xenograft models, patient-derived xenograft models, and glioblastoma organoids (GBOs). Single-cell RNA sequencing and mass cytometry were used to determine the immune phenotypes of CAR-T cells. We showed that locoregionally administered Tanζ-T28-Δ7R CAR-T cells induced long-term tumor regression with the desired safety outcomes. Patient-derived autologous Tanζ-T28-Δ7R CAR-T cells showed robust antitumor activity against GBOs. Our pre-clinical data has demonstrated the translational potential of Tanζ-T28-Δ7R CAR-T cell against GBM.
Collapse
Affiliation(s)
- You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China.
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Changqing Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Mingchen Yu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Zhongfang Shi
- Department of Pathophysiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China; China National Clinical Research Center for Neurological Diseases, Beijing, PR China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, PR China; Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, PR China; Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, PR China; Beijing Engineering Research Center of Targeted Drugs and Cell Therapy for CNS Tumors, Beijing, PR China.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China; China National Clinical Research Center for Neurological Diseases, Beijing, PR China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, PR China; Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, PR China; Beijing Engineering Research Center of Targeted Drugs and Cell Therapy for CNS Tumors, Beijing, PR China.
| |
Collapse
|
10
|
Riegel DC, Bureau BL, Conlon P, Chavez G, Connelly JM. Long-term survival, patterns of progression, and patterns of use for patients with newly diagnosed glioblastoma treated with or without Tumor Treating Fields (TTFields) in a real-world setting. J Neurooncol 2025:10.1007/s11060-025-04946-w. [PMID: 40163248 DOI: 10.1007/s11060-025-04946-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/17/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE Tumor Treating Fields therapy (TTFields) is an FDA-approved locoregional treatment for patients with newly diagnosed glioblastoma (ndGBM). Previous trial data showed the addition of TTFields to standard TMZ-based therapy to significantly improve overall survival (OS), but real-world data is lacking, particularly with long follow-up duration. Here, we report real-world survival, patterns of progression, and patterns of use for patients for patients with ndGBM treated with or without TTFields. METHODS Patients diagnosed with GBM and treated with standard of care therapy at the Medical College of Wisconsin between March 2015-March 2023 were included. Survival outcomes were assessed and compared across groups who received or did not receive TTFields therapy during maintenance treatment. Patients were followed through March 1, 2024. RESULTS A total of 208 patients (TTFields: n = 109; No-TTFields: n = 99) were included for analysis. Baseline characteristics were consistent across groups. Median OS and PFS were significantly improved for the TTFields group vs. No-TTFields group (median OS: 21.7 vs. 17.7 months, p = 0.029; median PFS: 12.4 vs. 9.6 months, p = 0.047). Patients treated with TTFields exhibited a higher rate of non-local progression vs. No-TTFields group. Median OS and PFS were each significantly longer for the ≥ 75% usage group compared with < 75% via matched analysis. CONCLUSION The results of this study reveal an association between TTFields use and long-term survival benefit, consistent with pivotal trial findings. TTFields use is associated with a higher incidence of non-local patterns of progression, and TTFields device usage ≥ 75% is associated with increased progression-free and long-term survival.
Collapse
Affiliation(s)
- Devon C Riegel
- Department of Neurology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Britta L Bureau
- Department of Neurology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | | | | | - Jennifer M Connelly
- Department of Neurology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| |
Collapse
|
11
|
Słychan K, Piersiak M, Rubin J, Kozioł A, Tyliszczak M, Pawłowski M, Chojak R. Regional and systemic complications following glioma resection: a systematic review and meta-analysis. Neurosurg Rev 2025; 48:323. [PMID: 40138052 DOI: 10.1007/s10143-025-03478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Gliomas represent a heterogeneous group of primary brain tumors with variable biological behavior. High-grade variants, notably glioblastomas, exhibit aggressive growth and a poor prognosis. Although surgical resection is central to management, it may lead to systemic and regional postoperative complications that adversely affect outcomes. While neurological complications have been extensively studied, comprehensive analyses of non-neurological sequelae remain limited. This study aimed to estimate the pooled proportion of postoperative systemic and regional complications following glioma resection and to identify factors influencing these proportions. A systematic review and meta-analysis was conducted per PRISMA guidelines. PubMed, Web of Science, and Embase were searched for English-language articles published between January 2000 and November 2024 reporting postoperative complications in glioma resection patients. Pooled proportion were calculated using a random-effects model, and meta-regression assessed the impact of covariates including patient age, gender, publication date, geographical location, and sample size. Seventy-seven studies were included. The pooled proportions for postoperative complications were as follows: venous thromboembolism, 4.92% (95% CI: 1.51-10.05%); deep vein thrombosis, 4.75% (95% CI: 2.86-7.05%); urinary tract infection, 3.77% (95% CI: 0.81-8.47%); hydrocephalus, 2.53% (95% CI: 1.37-3.97%); pulmonary infection, 2.39% (95% CI: 1.15-3.99%); cerebrospinal fluid leak, 2.22% (95% CI: 0.99-3.87%); surgical site infection, 2.21% (95% CI: 1.48-3.07%); meningitis, 1.49% (95% CI: 0.47-2.96%); pulmonary embolism, 1.33% (95% CI: 0.74-2.06%); and sepsis, 1.12% (95% CI: 0.08-3.02%). Significant heterogeneity was observed across studies, with meta-regression revealing that geographical location, publication date, and patient age were significant moderators influencing certain complication rates. This meta-analysis demonstrates that, while systemic and regional complication rates following glioma resection are relatively low, they remain clinically significant. In particular, venous thromboembolism and deep vein thrombosis are notably prevalent. Moreover, significant heterogeneity-shaped by geography, publication date, and patient age-underscores the need for tailored perioperative strategies and further research.
Collapse
Affiliation(s)
- Katarzyna Słychan
- Department of Neurosurgery, 4th Military Hospital in Wroclaw, Wrocław, Poland
| | - Marcin Piersiak
- Department of Neurosurgery, 4th Military Hospital in Wroclaw, Wrocław, Poland
| | - Jakub Rubin
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Aleksandra Kozioł
- Faculty of Dentistry, Wroclaw Medical University, Wroclaw, Poland
- Jan Mikulicz-Radecki University Clinical Hospital, Wroclaw, Poland
| | - Michał Tyliszczak
- Department of Pharmacology, Wroclaw Medical University, Wrocław, Poland
| | - Mateusz Pawłowski
- Department of Neurosurgery, St. Hedwig's Regional Specialist Hospital, Wodociągowa 4, Opole, 45-221, Poland
- Department of Neurosurgery, Institute of Medical Sciences, University of Opole, Al.Witosa 26, Opole, 45-401, Poland
| | - Rafał Chojak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
12
|
Okamoto T, Mizuta R, Demachi-Okamura A, Muraoka D, Sasaki E, Masago K, Yamaguchi R, Teramukai S, Otani Y, Date I, Tanaka S, Takahashi Y, Hashimoto N, Matsushita H. Immune prognostic model for glioblastoma based on the ssGSEA enrichment score. Cancer Genet 2025; 294-295:32-41. [PMID: 40121844 DOI: 10.1016/j.cancergen.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE Few effective immune prognostic models based on the tumor immune microenvironment (TIME) for glioblastoma have been reported. Therefore, this study aimed to construct an immune prognostic model for glioblastoma by analyzing enriched biological processes and pathways in tumors. METHODS A comprehensive single-sample gene set enrichment analysis (ssGSEA) of gene sets from the Molecular Signatures Database was performed using TCGA RNA sequencing data (141 glioblastoma cases). After evaluating gene sets associated with prognosis using univariable Cox regression, gene sets related to biological processes and tumor immunity in gliomas were extracted. Finally, the least absolute shrinkage and selection operator Cox regression refined the gene sets and a nomogram was constructed. The model was validated using CGGA (183 cases) and Aichi Cancer Center (42 cases) datasets. RESULTS The immune prognostic model consisted of three gene sets related to biological processes (sphingolipids, steroid hormones, and intermediate filaments) and one related to tumor immunity (immunosuppressive chemokine pathways involving tumor-associated microglia and macrophages). Kaplan-Meier curves for the training (TCGA) and validation (CGGA) cohorts showed significantly worse overall survival in the high-risk group compared to the low-risk group (p < 0.001 and p = 0.04, respectively). Furthermore, in silico cytometry revealed a significant increase in macrophages with immunosuppressive properties and T cells with effector functions in the high-risk group (p < 0.01) across all cohorts. CONCLUSION Construction of an immune prognostic model based on the TIME assessment using ssGSEA could potentially provide valuable insights into the prognosis and immune profiles of patients with glioblastoma and guide treatment strategies.
Collapse
Affiliation(s)
- Takanari Okamoto
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan; Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan.
| | - Ryo Mizuta
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan; Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Daisuke Muraoka
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Eiichi Sasaki
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Satoshi Teramukai
- Department of Biostatistics, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shota Tanaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshinobu Takahashi
- Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Naoya Hashimoto
- Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| |
Collapse
|
13
|
Holman R, McDannold N. Identifying new therapeutics for focused ultrasound-enhanced drug delivery in the management of glioblastoma. Front Oncol 2025; 15:1507940. [PMID: 40182047 PMCID: PMC11965939 DOI: 10.3389/fonc.2025.1507940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/07/2025] [Indexed: 04/05/2025] Open
Abstract
Glioblastoma, a grade IV astrocytoma, typically has a poor prognosis, with most patients succumbing within eighteen months of diagnosis and few experiencing long-term survival. Focused ultrasound, an emerging localized therapy, has shown promising results in early-phase studies for glioblastoma by improving the uptake of temozolomide and carboplatin. The blood-brain barrier is critical to homeostasis by regulating the movement of substances between the bloodstream and the central nervous system. While this barrier helps prevent infections from bloodborne pathogens, it also hinders the delivery of cancer therapies to gliomas. Combining focused ultrasound with circulating microbubbles enhances local blood-brain barrier permeability, facilitating the intratumoral uptake of systemic cancer therapies. The purpose of this study was to identify promising new therapeutics in the treatment of glioblastoma for localized drug delivery via focused ultrasound. This review provides an overview of the current standard of care for newly diagnosed and recurrent glioblastoma, identifies current therapies indicated for the treatment, discusses key aspects of microbubble resonators, describes focused ultrasound devices under evaluation in human trials, and concludes with a perspective of emerging therapeutics for future studies.
Collapse
Affiliation(s)
- Ryan Holman
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women’s Hospital, Boston, MA, United States
| | | |
Collapse
|
14
|
Brenner AJ, Patel T, Bao A, Phillips WT, Michalek JE, Youssef M, Weinberg JS, Kamiya Matsuoka C, Hedrick MH, LaFrance N, Moore M, Floyd JR. Convection enhanced delivery of Rhenium ( 186Re) Obisbemeda ( 186RNL) in recurrent glioma: a multicenter, single arm, phase 1 clinical trial. Nat Commun 2025; 16:2079. [PMID: 40055350 PMCID: PMC11889265 DOI: 10.1038/s41467-025-57263-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Rhenium (186Re) Obisbemeda (186RNL), chelated-186Re encapsulated in nanoliposomes and delivered to brain tumors via convection enhanced delivery (CED), was evaluated in a Phase 1 dose escalation trial (NCT01906385). The primary objective was to determine the maximum tolerated dose (MTD). Secondary objectives included safety and tolerability, dose distribution, the overall response rate (ORR), disease-specific progression-free survival (PFS), and overall survival (OS). 21 patients received up to 22.3 mCi 186RNL over 6 dosing cohorts. Most adverse events (AEs) were unrelated to 186RNL and the MTD was not reached. Although not predefined outcomes, the mOS and mPFS were 11 and 4 months, respectively, and found to correlate with radiation absorbed dose to the tumor and percent tumor treated. When dichotomized by absorbed dose of 100 Gy, the mOS and mPFS were 17 months and 6 months, respectively, for >100 Gy, compared to 6 (mOS) and 2 (mPFS) months, respectively, for <100 Gy. For ORR, 57.1% exhibited stable disease (SD), 4.8% partial response, and 38.1% progressive disease. Overall, patients received radiation absorbed doses without significant toxicity higher than possible with external beam radiation therapy (EBRT) and demonstrated mOS beyond standard of care for recurrent glioblastoma (~8 months).
Collapse
Affiliation(s)
- Andrew J Brenner
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA.
| | - Toral Patel
- UT Southwestern Medical Center of Dallas, Dallas, TX, USA
| | - Ande Bao
- Case Western Reserve University, Cleveland, OH, USA
| | | | - Joel E Michalek
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | | - John R Floyd
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
15
|
Jiang Y, Huang X, Huang R, Deng K, Dai L, Wang B. Prognostic modeling of disulfidptosis gene-associated lncRNAs aids in identifying the tumor microenvironment and guiding the selection of therapy. Discov Oncol 2025; 16:273. [PMID: 40053203 DOI: 10.1007/s12672-025-02033-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 03/03/2025] [Indexed: 03/10/2025] Open
Abstract
INTRODUCTION Gliomas, a type of malignant tumor, are marked by a short survival period and an unfavorable prognosis. Disulfide stress, which arises from an overabundance of intracellular cystine, can initiate disulfidoptosis, an emerging form of cell death. The link between gliomas and disulfidoptosis has not been extensively explored. This study breaks new ground by investigating the correlation between glioma prognosis and lncRNAs associated with disulfidoptosis, with the aim of improving glioma treatment strategies. METHODS We analyzed 10 long non-coding RNAs (lncRNAs) co-expressed with disulfidoptosis genes, retrieved clinical information and gene expression profiles from glioma and normal groups in the TCGA database, and developed a prognostic model for lncRNAs based on this data. The receiver operating characteristic curve (ROC) was used to evaluate and validate the model's reliability. Furthermore, the Kaplan-Meier survival curve was employed to assess the disparity in overall survival (OS) among patients with varying risk scores. We also examined the tumor microenvironment (TME), immune cell infiltration, immune-related functions, tumor mutational burden (TMB), and OncoPredict in samples with differing risk scores. To confirm the expression variations of genes associated with prognostic models in cell lines, quantitative polymerase chain reaction (qPCR) was employed. RESULTS Eleven long non-coding RNAs (lncRNAs) were identified for constructing prognostic models by analyzing lncRNAs associated with disulfidoptosis genes using Cox regression and LASSO regression analyses. The study's findings indicate that these 11 key lncRNAs serve as independent predictors of overall survival (OS) in glioma patients. Moreover, the frequency with which patients of varying risk scores opt for immune checkpoint blockade (ICB) therapy and chemotherapy not only differs but also their responses to these treatments are significantly distinct, suggesting that the risk score could be a predictive factor for treatment response. CONCLUSIONS This research sheds light on the characteristics of disulfidoptosis in glioma, revealing that patterns of disulfidoptosis in patients can be effectively assessed using a risk score. Consequently, the judicious application of this prognostic model can significantly inform clinical treatment strategies and precision medicine for glioma, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Ying Jiang
- Cerebrovascular Diseases Center, Department of Neurosurgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Xueping Huang
- Department of Neurology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Rong Huang
- Department of Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Kaihan Deng
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Lin Dai
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Bin Wang
- Department of Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
16
|
Kuroda H, Kijima N, Tachi T, Ikeda S, Murakami K, Nakagawa T, Yaga M, Nakagawa K, Utsugi R, Hirayama R, Okita Y, Kagawa N, Hosen N, Kishima H. Prostaglandin F2 receptor negative regulator as a potential target for chimeric antigen receptor-T cell therapy for glioblastoma. Cancer Immunol Immunother 2025; 74:136. [PMID: 40047938 PMCID: PMC11885767 DOI: 10.1007/s00262-025-03979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapy targeting novel glioblastoma (GBM)-specific cell surface antigens is a promising approach. However, transcriptome analyses have revealed few GBM-specific target antigens. METHODS A library of monoclonal antibodies (mAbs) against tumor cell lines derived from patients with GBM was generated. mAbs reacting with tumor cells in resected tissues from patients with GBM but not with nonmalignant human brain cells were detected. The antigens that were recognized were identified through expression cloning. CAR-T cells derived from a candidate mAb were generated, and their functionality was tested in vitro and in vivo. RESULTS Approximately 3,200 clones were established. Among them, 5E17 reacted with tumor cells in six of seven patients with GBM, but not with nonmalignant human brain cells. Prostaglandin F2 receptor negative regulator (PTGFRN) was identified as an antigen recognized by 5E17. CAR-T cells derived from 5E17 produced cytokines and exerted cytotoxicity upon co-culture with tumor cells from patients with GBM. Furthermore, intracranial injection of 5E17-CAR-T cells demonstrated antitumor effects in an orthotopic xenograft murine model with patient-derived GBM cells. CONCLUSIONS Cell surface PTGFRN is a candidate target for intracranial CAR-T cell therapy for GBM. On-target off-tumor toxicity in alternative normal tissues needs to be carefully tested.
Collapse
Affiliation(s)
- Hideki Kuroda
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Noriyuki Kijima
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan.
| | - Tetsuro Tachi
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Shunya Ikeda
- World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Koki Murakami
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Tomoyoshi Nakagawa
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Moto Yaga
- Department of Respiratory Medicine, Osaka General Hospital, Osaka, Osaka, Japan
| | - Kanji Nakagawa
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Reina Utsugi
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Ryuichi Hirayama
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Yoshiko Okita
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Naoki Kagawa
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| | - Naoki Hosen
- World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan.
| | - Haruhiko Kishima
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, 5650871, Japan
| |
Collapse
|
17
|
Cheng Y, Xiao Z, Cai W, Zhou T, Yang Z. Suppression of FOXO1 activity by SIRT1-mediated deacetylation weakening the intratumoral androgen autocrine function in glioblastoma. Cancer Gene Ther 2025; 32:343-354. [PMID: 40075208 PMCID: PMC11946903 DOI: 10.1038/s41417-025-00880-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025]
Abstract
Elevated levels of androgens in the brain accelerate tumor progression in patients with glioblastoma (GBM). Despite current research efforts concentrating on decreasing peripheral androgens to improve GBM prognosis, results have not met expectations. Herein, we aim to elucidate the source of increased androgen levels in the brains of GBM patients and investigate whether lowering it can improve the prognosis of GBM patients. The Elisa was employed to measure androgen levels. The effects of androgens on U87 cells were evaluated using CCK-8 assays, clone formation assays, wound healing assays, and migration/invasion assays. RNA sequencing, RT-qPCR and Western blotting were performed to assess the expression levels of steroid enzymes, tumor drug resistance, Sirt1, FOXO1genes and proteins. Co-immunoprecipitation (Co-IP) assays were conducted to investigate the interactions and acetylation levels between Sirt1 and FOXO1. Lentiviral transfection was utilized to establish stable cell lines. Furthermore, an in vivo murine subcutaneous tumor model was established to further confirm the role of Sirt1 in tumor progression. We found androgen levels in the cerebrospinal fluid of GBM patients were higher than in the periphery, contrasting with healthy individuals. Additionally, the steroid enzymes in GBM cells were upregulated. Reducing peripheral androgens compensatorily enhances GBM androgen synthesis capacity (CYP17A1, CYP11A1, SRD5A2) and chemo-resistance (ABCB11, BIRC3, FGF2, NRG1), while the levels of androgens in the brain remain consistently high. The above results indicate that the increased androgens in the brain of GBM patients are self-secreted. Further investigations demonstrate that the transcription factor FOXO1 in GBM is regulated by silent information regulator 1 (Sirt1) through deacetylation, leading to enhanced androgen synthesis capacity in vivo and in vitro. Overexpressing Sirt1 significantly lowers brain androgen levels and delays tumor progression in mouse models. Compared to conventional finasteran therapy, the targeted-Sirt1 results in lower brain androgen levels and smaller tumor volumes. Our findings provide evidence that the elevated androgens in the brain of GBM patients came from tumor autocrine. Overexpression of Sirt1 reduces FOXO1 acetylation, lowers androgen synthesis enzyme levels, and effectively decreases brain androgen levels, thereby delaying tumor progression.
Collapse
Affiliation(s)
- Yuanchi Cheng
- Department of Neurosurgery, Shanghai University of Medicine & Health Science Affiliated Sixth People's Hospital South Campus, Shanghai, 201499, China
| | - Zhijun Xiao
- Department of Pharmacy, Shanghai University of Medicine & Health Science Affiliated Sixth People's Hospital South Campus, Shanghai, 201499, China
| | - Weijia Cai
- Department of Pharmacy, Shanghai University of Medicine & Health Science Affiliated Sixth People's Hospital South Campus, Shanghai, 201499, China
| | - Ting Zhou
- Department of Pharmacy, Shanghai University of Medicine & Health Science Affiliated Sixth People's Hospital South Campus, Shanghai, 201499, China.
| | - Zhen Yang
- Department of Central Laboratory, Shanghai University of Medicine & Health Science Affiliated Sixth People's Hospital South Campus, Shanghai, 201499, China.
| |
Collapse
|
18
|
Fathi Kazerooni A, Akbari H, Hu X, Bommineni V, Grigoriadis D, Toorens E, Sako C, Mamourian E, Ballinger D, Sussman R, Singh A, Verginadis II, Dahmane N, Koumenis C, Binder ZA, Bagley SJ, Mohan S, Hatzigeorgiou A, O'Rourke DM, Ganguly T, De S, Bakas S, Nasrallah MP, Davatzikos C. The radiogenomic and spatiogenomic landscapes of glioblastoma and their relationship to oncogenic drivers. COMMUNICATIONS MEDICINE 2025; 5:55. [PMID: 40025245 PMCID: PMC11873127 DOI: 10.1038/s43856-025-00767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/12/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glioblastoma is a highly heterogeneous brain tumor, posing challenges for precision therapies and patient stratification in clinical trials. Understanding how genetic mutations influence tumor imaging may improve patient management and treatment outcomes. This study investigates the relationship between imaging features, spatial patterns of tumor location, and genetic alterations in IDH-wildtype glioblastoma, as well as the likely sequence of mutational events. METHODS We conducted a retrospective analysis of 357 IDH-wildtype glioblastomas with pre-operative multiparametric MRI and targeted genetic sequencing data. Radiogenomic signatures and spatial distribution maps were generated for key mutations in genes such as EGFR, PTEN, TP53, and NF1 and their corresponding pathways. Machine and deep learning models were used to identify imaging biomarkers and stratify tumors based on their genetic profiles and molecular heterogeneity. RESULTS Here, we show that glioblastoma mutations produce distinctive imaging signatures, which are more pronounced in tumors with less molecular heterogeneity. These signatures provide insights into how mutations affect tumor characteristics such as neovascularization, cell density, invasion, and vascular leakage. We also found that tumor location and spatial distribution correlate with genetic profiles, revealing associations between tumor regions and specific oncogenic drivers. Additionally, imaging features reflect the cross-sectionally inferred evolutionary trajectories of glioblastomas. CONCLUSIONS This study establishes clinically accessible imaging biomarkers that capture the molecular composition and oncogenic drivers of glioblastoma. These findings have potential implications for noninvasive tumor profiling, personalized therapies, and improved patient stratification in clinical trials.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Xiaoju Hu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Vikas Bommineni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitris Grigoriadis
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Erik Toorens
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Ballinger
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robyn Sussman
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nadia Dahmane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Artemis Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tapan Ganguly
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - MacLean P Nasrallah
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Weng X, Gonzalez M, Angelia J, Piroozmand S, Jamehdor S, Behrooz AB, Latifi-Navid H, Ahmadi M, Pecic S. Lipidomics-driven drug discovery and delivery strategies in glioblastoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167637. [PMID: 39722408 DOI: 10.1016/j.bbadis.2024.167637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
With few viable treatment options, glioblastoma (GBM) is still one of the most aggressive and deadly types of brain cancer. Recent developments in lipidomics have demonstrated the potential of lipid metabolism as a therapeutic target in GBM. The thorough examination of lipids in biological systems, or lipidomics, is essential to comprehending the changed lipid profiles found in GBM, which are linked to the tumor's ability to grow, survive, and resist treatment. The use of lipidomics in drug delivery and discovery is examined in this study, focusing on how it may be used to find new biomarkers, create multi-target directed ligands, and improve drug delivery systems. We also cover the use of FDA-approved medications, clinical trials that use lipid-targeted medicines, and the integration of lipidomics with other omics technologies. This study emphasizes lipidomics as a possible tool in developing more effective treatment methods for GBM by exploring various lipid-centric techniques.
Collapse
Affiliation(s)
- Xiaohui Weng
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Michael Gonzalez
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Jeannes Angelia
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States.
| |
Collapse
|
20
|
Guo Y, Jin L, Shen Z, Fan L, Yu X, Kuang Y, Cai L, Zhou J, Chen Z, Yan F, Zhang J, Tong M, Yuan J, Mao Z, Chen G. Biomimetic Membrane Vesicles Reprogram Microglia Polarization and Remodel the Immunosuppressive Microenvironment of Glioblastoma via PERK/HIF-1α/Glycolysis Pathway. Adv Healthc Mater 2025; 14:e2404782. [PMID: 39757442 DOI: 10.1002/adhm.202404782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Indexed: 01/07/2025]
Abstract
The malignant interaction between tumor cells and immune cells is one of the important reasons for the rapid progression and refractoriness of glioblastoma (GBM). As an essential metabolic center of M2 macrophages, the inhibition of protein kinase RNA-like endoplasmic reticulum kinase (PERK) leads to the reduction of M2 macrophages. Nevertheless, the restriction of the blood-brain barrier (BBB) and non-specific cell targeting hinder the application of PERK inhibitors in GBM. Herein, the optimal NP-M-M2pep is developed successfully, which has shown the capacity of BBB penetration and specific targeting of M2 microglia. In addition to inhibiting the polarization of M2 microglia, the administration of iPERK@NP-M-M2pep reprogrammed M2 microglia into M1 ones in vitro via PERK/HIF-1α/glycolysis pathway. Efficient brain accumulation of nanoparticles is achieved after tail vein injection, with effective inhibition of GBM progression after one course of treatment. The glioma-associated microglia and macrophages (GAM) with M2 type are induced to M1 and the immunosuppressive TME is remodeled by upregulating immunostimulatory cells and downregulating immunosuppressive cells. In summary, the biomimetic membrane vesicles (BMVs) specifically delivered iPERK to GAMs offer an inspiring strategy to reprogram microglia polarization, re-educate immunosuppressive TME, and inhibit the progression of GBM.
Collapse
Affiliation(s)
- Yinghan Guo
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhipeng Shen
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Linfeng Fan
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Xian Yu
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Yirui Kuang
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Lingxin Cai
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Jiayin Zhou
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Zihang Chen
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Feng Yan
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Jianmin Zhang
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| | - Minfeng Tong
- Department of Neurosurgery, affiliated Jinhua Hospital, School of Medicine, Zhejiang University, Jinhua, Zhejiang, 321000, China
| | - Jianlie Yuan
- Department of Neurosurgery, affiliated Jinhua Hospital, School of Medicine, Zhejiang University, Jinhua, Zhejiang, 321000, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Gao Chen
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Key Laboratory of Research and Transformation for Major Neurosurgical Diseases, Hangzhou, Zhejiang, 310009, China
| |
Collapse
|
21
|
Lan CW, Chen HH, Sheu JJC. Deoxyelephantopin induces apoptosis and cell cycle arrest in GL261 glioblastoma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2729-2738. [PMID: 39254878 DOI: 10.1007/s00210-024-03429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant central nervous system tumor with a poor prognosis. Developing new therapeutic drugs is crucial. This study evaluates deoxyelephantopin (DET), a major component of *Elephantopus scaber* L., for its potential anti-GBM effects. The effects of DET on GBM cell lines were investigated using the MTT assay and Annexin-V kit to assess cell death and apoptosis. Western blot analysis examined apoptosis and cell cycle-related proteins. ELISA kits measured VEGF and TGF-β levels. In vivo, NOD SCID mice were injected with GL-261 cells and treated with DET to evaluate tumor growth and survival. DET inhibited GBM cell growth in a time- and dose-dependent manner. MTT and Annexin-V assays confirmed cell death and apoptosis. Western blot analysis showed DET downregulated Bcl-2 and increased caspase-3, Bax, and cytochrome c levels. ELISA results indicated that DET suppressed VEGF and TGF-β expression. DET treatment also decreased phosphorylation of AKT and STAT-3, CDK4, cyclin D2, MMP2, and MMP9 levels. In vivo, DET significantly inhibited tumor growth and improved survival rates in mice. DET exhibits significant in vitro and in vivo anticancer effects, making it a promising candidate for further research and potential clinical application against GBM.
Collapse
Affiliation(s)
- Chun-Wen Lan
- Institute of Biomedical Sciences, National Sun Yat-sen University, No.70 Lien-hai Road, Kaohsiung, 804201, Taiwan
| | - Hsin-Hung Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, No. 386, Dazhong 1st Rd., Zuoying Dist., Kaohsiung City, 813414, Taiwan.
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, No.70 Lien-hai Road, Kaohsiung, 804201, Taiwan.
- School of Chinese Medicine, China Medical University, Taichung, 404333, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.
| |
Collapse
|
22
|
Vallieri N, Datsi A. Immune Cell Interplay in the Fight Against GBM. Cancers (Basel) 2025; 17:817. [PMID: 40075663 PMCID: PMC11899300 DOI: 10.3390/cancers17050817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Despite multimodal therapies, the treatment of glioblastoma remains challenging. In addition to the very complex mechanisms of cancer cells, including specialized phenotypes that enable them to proliferate, invade tissues, and evade immunosurveillance, they exhibit a pronounced resistance to chemo- and radiotherapy. More advanced tumors create a hypoxic environment that supports their proliferation and survival, while robust angiogenesis ensures a constant supply of nutrients. In GBM, these structures are very pronounced and contribute to the creation and maintenance of a highly immunosuppressive microenvironment that promotes tumor growth and immune escape. In addition, the high accumulation of immunosuppressive tumor-infiltrating leukocytes and other cells, the pronounced expression of immune checkpoint molecules, and the low mutational burden, i.e., the low number of neoantigens, are hallmarks of GBM and contribute to the challenge of therapeutic approaches. Here, we review a number of mechanisms that GBM exploits to support tumor growth and potential treatments. These include new chemotherapeutics, tumor treating fields, and small molecules, including compounds targeting angiogenesis or blockers of tyrosine kinases that inhibit tumor cell proliferation and survival. In addition, we focus on immunotherapies such as immune checkpoint blockade or cell therapies, in particular vaccination with dendritic cells and CAR-T cells, which can either kill GBM cells directly or bypass immunosuppression by modulating the tumor microenvironment or boosting the patient's own immune response.
Collapse
Affiliation(s)
| | - Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, 40225 Duesseldorf, Germany;
| |
Collapse
|
23
|
Nisnboym M, Sneiderman CT, Jaswal AP, Xiong Z, Vincze SR, Sever RE, Zou H, Frederico SC, Agnihotri S, Hu B, Drappatz J, Pollack IF, Kohanbash G, Raphael I. Assessment of anti-CD69 antibody therapy alone or in combination with anti-PD-1 in murine GBM. Expert Rev Clin Immunol 2025; 21:239-247. [PMID: 39402706 DOI: 10.1080/1744666x.2024.2412770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/27/2024] [Indexed: 02/02/2025]
Abstract
BACKGROUND Glioblastoma (GBM) is an aggressive cancer with limited treatment options. Immunotherapy targeting CD69, an early activation marker on T cells, has shown promise in preclinical models of non-CNS malignancies. This study investigates anti-CD69 therapy alone or in combination with anti-PD-1 in a preclinical GBM model. RESEARCH DESIGN AND METHODS CD69 expression in GBM patient tissues was analyzed using the TCGA database. Therapeutic efficacy of anti-CD69 was tested in a murine GBM model with different regimens. Immune cell populations in the tumor microenvironment (TME) were assessed by flow cytometry. RESULTS Increased CD69 expression was observed in GBM patients compared to normal brain tissue and was associated with worse prognosis. Anti-CD69 treatment reduced percentages of CD69+ immune cells but did not improve survival in GBM-bearing mice. Increased PD-1 expression on NK cells was observed following anti-CD69 treatment. Anti-CD69 treatment was not improved by the addition of anti-PD-1 in vivo. CONCLUSIONS This is the first study evaluating anti-CD69 therapy in a preclinical GBM model. Despite promising preclinical data in other cancers, anti-CD69 monotherapy or combination therapy with anti-PD-1 did not improve survival in this GBM model.
Collapse
MESH Headings
- Animals
- Glioblastoma/immunology
- Glioblastoma/drug therapy
- Glioblastoma/therapy
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Mice
- Lectins, C-Type/immunology
- Lectins, C-Type/antagonists & inhibitors
- Lectins, C-Type/metabolism
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Humans
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Brain Neoplasms/immunology
- Brain Neoplasms/drug therapy
- Brain Neoplasms/therapy
- Immunotherapy/methods
- Tumor Microenvironment/immunology
- Disease Models, Animal
- Killer Cells, Natural/immunology
- Cell Line, Tumor
- Immune Checkpoint Inhibitors/therapeutic use
- Female
Collapse
Affiliation(s)
- Michal Nisnboym
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv, Israel
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Chaim T Sneiderman
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ambika P Jaswal
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zujian Xiong
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah R Vincze
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - ReidAnn E Sever
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Han Zou
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen C Frederico
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Baoli Hu
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jan Drappatz
- Departments of Neurology and Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ian F Pollack
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Itay Raphael
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
24
|
Fortunato JT, Walsh LE, Polacek LC, Reiner AS, Walbert T, Thomas AA, Buthorn J, Sigler A, Prigerson HG, Applebaum AJ, Diamond EL. Illness understanding and religiousness in patients with recurrent glioblastoma. Neurooncol Pract 2025; 12:100-112. [PMID: 39917760 PMCID: PMC11798605 DOI: 10.1093/nop/npae068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2025] Open
Abstract
Background Patients with glioblastoma (GBM) often have inaccurate perceptions of prognosis. Strong religious beliefs have been associated with limited illness understanding (IU) in patients with advanced cancer, but IU and religiousness have not been investigated in patients with GBM. The aim of this study was to evaluate the association between religiousness and spirituality and IU in patients with GBM. Methods Patients enrolled in a prospective multicenter study of recurrent GBM (Coping with Glioblastoma, NCT02375841). Within one month of medical visits discussing MRI scans showing GBM progression, patients completed study surveys containing published measures of IU and religiousness. IU was compared between participants with moderate or high versus slight or no religiousness based on several patient-reported prompts using Fisher's exact tests. Results Twenty-four patients completed surveys of religiousness and IU. IU was partial within our cohort. Fifteen participants (62.5%) acknowledged that their illness was terminal. Only 6 (25%) correctly acknowledged their prognosis (months). Eleven patients (46%) were moderately or very religious, while 9 (38%) were either slightly religious or not at all religious. High religiousness and spirituality were each associated with partial IU (P = .06 and P = .01, respectively). A belief that God could perform a miracle to cure them of cancer and a belief in sanctity through suffering were also each associated with partial IU. Conclusions This prospective study that suggests religiousness, including the belief in miracles and a belief in sanctification through suffering, might influence patients' IU. Further research is warranted to study this association.
Collapse
Affiliation(s)
- John T Fortunato
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Leah E Walsh
- Department of Psychology, Fordham University, Bronx, New York, USA
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Laura C Polacek
- Department of Psychology, Fordham University, Bronx, New York, USA
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Anne S Reiner
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tobias Walbert
- Department of Neurosurgery and Neurology, Henry Ford Health, Wayne State and Michigan State University, Detroit, Michigan, USA
| | - Alissa A Thomas
- Department of Neurological Sciences, Larner College of Medicine at the University of Vermont, Burlington, Vermont, USA
| | - Justin Buthorn
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Allison Sigler
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Holly G Prigerson
- Cornell Center for Research on End of Life Care, Department of Radiology and Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Allison J Applebaum
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Eli L Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
25
|
Juarez TM, Gill JM, Minev BR, Sharma A, Kesari S. Neoadjuvant clinical trials in adults with newly diagnosed high-grade glioma: A systematic review. Crit Rev Oncol Hematol 2025; 206:104596. [PMID: 39675399 DOI: 10.1016/j.critrevonc.2024.104596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND High-grade gliomas are devastating cancers that remain incurable with standard surgical resection and radiochemotherapy. Although beneficial against neoplasms, radiation lowers lymphocyte counts, weakens immune activation, and recruits suppressive myeloid cells impairing immune responses. Tumor environments treated with radiation experience long-term immunosuppression, reducing immunotherapy effectiveness and contributing to recurrence. Investigating pre-radiation treatments in newly diagnosed patients could identify active agents, assess immunotherapy impact, and enable multiomic analyses without radiation-induced confounding factors. This literature review was conducted to describe the feasibility, safety, and outcomes of postsurgical, pre-radiation clinical trials for adults with newly diagnosed high-grade glioma. METHODS A systematic review was performed of the English-language literature reporting results of clinical trials for adults with newly diagnosed high-grade glioma administered postsurgical treatment prior to radiation therapy. A search was conducted in PubMed and references cited in research and review articles were also considered. RESULTS From 1991 to 2024, 52 clinical trials were identified: 3 phase I, 38 phase II, 4 phase III, and 7 of unknown phase. Nine trials were randomized, 24 were multicenter trials, 21 investigated temozolomide-containing regimens, and 12 focused on inoperable tumors, involving a total of 2737 patients. CONCLUSION Pre-radiation neoadjuvant studies are feasible and may identify active drugs. This is particularly relevant in the era of personalized medicine with brain-penetrant drugs, targeted therapy, and immuno-oncology advancements. Investigating pre-radiation treatments in newly diagnosed high-grade glioma is a viable approach to rapidly identify active and inactive regimens while the immune system and tumor microenvironment remain intact.
Collapse
Affiliation(s)
| | | | - Boris R Minev
- Calidi Biotherapeutics, San Diego, CA, USA; Department of Radiation Medicine and Applied Sciences, University of California, San Diego, CA 92093, USA
| | - Akanksha Sharma
- Pacific Neuroscience Institute, Santa Monica, CA, USA; Department of Translational Neuroscience, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute, Santa Monica, CA, USA; Department of Translational Neuroscience, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA.
| |
Collapse
|
26
|
Park SS, Roh TH, Tanaka Y, Kim YH, Park SH, Kim TG, Eom SY, Park TJ, Park IH, Kim SH, Kim JH. High p16 INK4A expression in glioblastoma is associated with senescence phenotype and better prognosis. Neoplasia 2025; 60:101116. [PMID: 39724755 PMCID: PMC11729681 DOI: 10.1016/j.neo.2024.101116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Glioblastoma, isocitrate dehydrogenase (IDH)-wildtype (GBM), is the most malignant brain tumor in adults, with limited therapeutic intervention. Previous studies have identified a few prognostic markers for GBM, including the methylation status of O6-methylguanine-DNA methyltransferase (MGMT) promoter, TERT promoter mutation, EGFR amplification, and CDKN2A/2B deletion. However, the classification of GBM remains incomplete, necessitating a comprehensive analysis. In this study, we investigated the impact of p16INK4A expression in GBM and found that p16INK4A-high GBM exhibits distinct characteristics compared to p16INK4A-low GBM. Specifically, tumor cells with p16INK4A-high expression display a senescent phenotype and are correlated with higher intra-tumoral immune cell infiltration. Furthermore, an association was observed between elevated p16INK4A expression in GBM and extended overall survival of patients. Our in vivo and in vitro studies revealed that CCL13 is predominantly expressed by p16INK4A-high GBM cells. The released CCL13 enhances the infiltration of T cells within the tumor, potentially contributing to the improved prognosis observed in patients with high p16INK4A expression. These findings suggest that tumor cells with a senescence phenotype in GBM, through the secretion of chemokines such as CCL13, may augment immune cell infiltration and potentially enhance patient outcomes by creating a more immunologically active tumor microenvironment.
Collapse
Affiliation(s)
- Soon Sang Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Inflammaging Translational Research Center, Ajou University Hospital, Suwon 16499, Republic of Korea
| | - Tae Hoon Roh
- Department of Neurosurgery, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Yoshiaki Tanaka
- Maisonneuve-Rosemont Hospital Research Center, Department of Medicine, University of Montreal, H1T2M4 Canada
| | - Young Hwa Kim
- Inflammaging Translational Research Center, Ajou University Hospital, Suwon 16499, Republic of Korea; Department of Pathology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - So Hyun Park
- Inflammaging Translational Research Center, Ajou University Hospital, Suwon 16499, Republic of Korea; Department of Pathology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Tae-Gyu Kim
- Inflammaging Translational Research Center, Ajou University Hospital, Suwon 16499, Republic of Korea; Department of Pathology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - So Yeong Eom
- Department of Pathology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Tae Jun Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Inflammaging Translational Research Center, Ajou University Hospital, Suwon 16499, Republic of Korea
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven 06520, USA
| | - Se-Hyuk Kim
- Department of Neurosurgery, Ajou University School of Medicine, Suwon 16499, Republic of Korea.
| | - Jang-Hee Kim
- Department of Pathology, Ajou University School of Medicine, Suwon 16499, Republic of Korea.
| |
Collapse
|
27
|
de Godoy LL, Rajan A, Banihashemi A, Patel T, Desai A, Bagley S, Brem S, Chawla S, Mohan S. Response Assessment in Long-Term Glioblastoma Survivors Using a Multiparametric MRI-Based Prediction Model. Brain Sci 2025; 15:146. [PMID: 40002479 PMCID: PMC11852837 DOI: 10.3390/brainsci15020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose: Early treatment response assessments are crucial, and the results are known to better correlate with prognosis and survival outcomes. The present study was conducted to differentiate true progression (TP) from pseudoprogression (PsP) in long-term-surviving glioblastoma patients using our previously established multiparametric MRI-based predictive model, as well as to identify clinical factors impacting survival outcomes in these patients. Methods: We report six patients with glioblastoma that had an overall survival longer than 5 years. When tumor specimens were available from second-stage surgery, histopathological analyses were used to classify between TP (>25% characteristics of malignant neoplasms; n = 2) and PsP (<25% characteristics of malignant neoplasms; n = 2). In the absence of histopathology, modified RANO criteria were assessed to determine the presence of TP (n = 1) or PsP (n = 1). The predictive probabilities (PPs) of tumor progression were measured from contrast-enhancing regions of neoplasms using a multiparametric MRI-based prediction model. Subsequently, these PP values were used to define each lesion as TP (PP ≥ 50%) or PsP (PP < 50%). Additionally, detailed clinical information was collected. Results: Our predictive model correctly identified all patients with TP (n = 3) and PsP (n = 3) cases, reflecting a significant concordance between histopathology/modified RANO criteria and PP values. The overall survival varied from 5.1 to 12.3 years. Five of the six glioblastoma patients were MGMT promoter methylated. All patients were female, with a median age of 56 years. Moreover, all six patients had a good functional status (KPS ≥ 70), underwent near-total/complete resection, and received alternative therapies. Conclusions: Multiparametric MRI can aid in assessing treatment response in long-term-surviving glioblastoma patients.
Collapse
Affiliation(s)
- Laiz Laura de Godoy
- Departments of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (S.M.)
| | - Archith Rajan
- Departments of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (S.M.)
| | - Amir Banihashemi
- Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Thara Patel
- Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (T.P.); (S.B.)
| | - Arati Desai
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (S.B.)
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen Bagley
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (S.B.)
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Brem
- Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (T.P.); (S.B.)
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (S.B.)
- Glioblastoma Translational Center of Excellence, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sanjeev Chawla
- Departments of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (S.M.)
| | - Suyash Mohan
- Departments of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (L.L.d.G.); (A.R.); (S.M.)
| |
Collapse
|
28
|
Giuffrida AS, Ramesh K, Sheriff S, Maudsley AA, Weinberg BD, Cooper LAD, Shim H. An Accelerated Spectroscopic MRI Metabolite Quantification Based on a Deep Learning Method for Radiation Therapy Planning in Brain Tumor Patients. Cancers (Basel) 2025; 17:423. [PMID: 39941791 PMCID: PMC11816355 DOI: 10.3390/cancers17030423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/08/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Spectroscopic MRI (sMRI) is a quantitative imaging technique that maps infiltrated tumors in the brain without contrast injections. In a previous study (NCT03137888), sMRI-guided radiation treatment extended patient survival, showing promise for clinical translation. The spectral fitting of individual voxels in an sMRI dataset generate metabolite concentration maps that guide treatment. The established spectral analysis methods use iterative least-squares fitting (FITT) that are computationally demanding. This study compares the performance of NNFit, a neural network-based, accelerated spectral fitting model, to the established FITT for metabolite quantification and radiation treatment planning. METHODS NNFit is a self-supervised deep learning model trained on 50 ms echo-time (TE) sMRI data to estimate metabolite levels of choline (Cho), creatine (Cr), and NAA. We trained the model on 30 GBM patients (56 scans) and tested it on 17 GBM patients (29 scans). NNFit's performance was compared to the FITT using structural similarity indices (SSIM) and the Dice coefficient. RESULTS NNFit significantly improved processing speed while maintaining strong agreement with FITT. The radiation target volumes defined by Cho/NAA ≥ 2x were visually comparable, with fewer artifacts in NNFit. Structural similarity indices (SSIM) indicated minimal bias and high consistency across methods. CONCLUSIONS This study highlights NNFit's potential for rapid, accurate, and artifact-reduced metabolic imaging, enabling faster radiotherapy planning.
Collapse
Affiliation(s)
- Alexander S. Giuffrida
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.S.G.); (K.R.)
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Karthik Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.S.G.); (K.R.)
| | - Sulaiman Sheriff
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 45056, USA; (S.S.); (A.A.M.)
| | - Andrew A. Maudsley
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 45056, USA; (S.S.); (A.A.M.)
| | - Brent D. Weinberg
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lee A. D. Cooper
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60657, USA;
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.S.G.); (K.R.)
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
29
|
Martinez-Paniagua M, Khan S, Henning NW, Konagalla SV, Patel CB. Optimized Methods to Quantify Tumor Treating Fields (TTFields)-Induced Permeabilization of Glioblastoma Cell Membranes. Methods Protoc 2025; 8:10. [PMID: 39997634 PMCID: PMC11858626 DOI: 10.3390/mps8010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
Glioblastoma (GBM) is a lethal primary brain cancer with a 5.6% five-year survival rate. Tumor treating fields (TTFields) are alternating low-intensity electric fields that have demonstrated a GBM patient survival benefit. We previously reported that 0.5-24 h of TTFields exposure resulted in an increased uptake of FITC-dextran fluorescent probes (4-20 kDa) in human GBM cells. However, this approach, in which a fluorescence plate-based detector is used to evaluate cells attached to glass coverslips, cannot distinguish FITC-dextran uptake in live vs. dead cells. The goal of the study was to report the optimization and validation of two independent methods to quantify human GBM cell membrane permeabilization induced by TTFields exposure. First, we optimized flow cytometry by measuring mean fluorescence intensity at 72 h for 4 kDa (TTFields 6726 ± 958.0 vs. no-TTFields 5093 ± 239.7, p = 0.016) and 20 kDa (7087 ± 1137 vs. 5055 ± 897.8, p = 0.031) probes. Second, we measured the ratio of lactate dehydrogenase (LDH) to cell viability (measured using the CellTiter-Glo [CTG] viability assay); the LDH/CTG ratio was higher under TTFields (1.47 ± 0.15) than no-TTFields (1.08 ± 0.08) conditions, p < 0.0001. The findings using these two independent methods reproducibly demonstrated their utility for time-dependent evaluations. We also showed that these methods can be used to relate the cell membrane-permeabilizing effects of the non-ionizing radiation of TTFields to that of an established cell membrane permeabilizer, the non-ionic detergent Triton-X-100. Evaluating carboplatin ± TTFields, the LDH/CTG ratio was significantly higher in the TTFields vs. no-TTFields condition at each carboplatin concentration (0-30 µM), p = 0.014. We successfully optimized and validated two cost-effective methods to reproducibly quantify TTFields-induced human GBM cancer cell membrane permeabilization.
Collapse
Affiliation(s)
- Melisa Martinez-Paniagua
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1002, BSRB S5.8116b, Houston, TX 77030, USA; (M.M.-P.); (S.K.); (N.W.H.)
| | - Sabbir Khan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1002, BSRB S5.8116b, Houston, TX 77030, USA; (M.M.-P.); (S.K.); (N.W.H.)
| | - Nikita W. Henning
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1002, BSRB S5.8116b, Houston, TX 77030, USA; (M.M.-P.); (S.K.); (N.W.H.)
| | - Sri Vaishnavi Konagalla
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1002, BSRB S5.8116b, Houston, TX 77030, USA; (M.M.-P.); (S.K.); (N.W.H.)
| | - Chirag B. Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1002, BSRB S5.8116b, Houston, TX 77030, USA; (M.M.-P.); (S.K.); (N.W.H.)
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center/The University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center/The University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
30
|
Yang L, Li S, Yu L, Leng J, Li N. Targeting glycolysis: exploring a new frontier in glioblastoma therapy. Front Immunol 2025; 15:1522392. [PMID: 39877360 PMCID: PMC11772265 DOI: 10.3389/fimmu.2024.1522392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Glioblastoma(GBM) is a highly malignant primary central nervous system tumor that poses a significant threat to patient survival due to its treatment resistance and rapid recurrence.Current treatment options, including maximal safe surgical resection, radiotherapy, and temozolomide (TMZ) chemotherapy, have limited efficacy.In recent years, the role of glycolytic metabolic reprogramming in GBM has garnered increasing attention. This review delves into the pivotal role of glycolytic metabolic reprogramming in GBM, with a particular focus on the multifaceted roles of lactate, a key metabolic product, within the tumor microenvironment (TME). Lactate has been implicated in promoting tumor cell proliferation, invasion, and immune evasion. Additionally, this review systematically analyzes potential therapeutic strategies targeting key molecules within the glycolytic pathway, such as Glucose Transporters (GLUTs), Monocarboxylate Transporters(MCTs), Hexokinase 2 (HK2), 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 3 (PFKFB3), Pyruvate Kinase Isozyme Type M2 (PKM2), and the Lactate Dehydrogenase A (LDHA). These studies provide a novel perspective for GBM treatment. Despite progress made in existing research, challenges remain, including drug penetration across the blood-brain barrier, side effects, and resistance. Future research will aim to address these challenges by improving drug delivery, minimizing side effects, and exploring combination therapies with radiotherapy, chemotherapy, and immunotherapy to develop more precise and effective personalized treatment strategies for GBM.
Collapse
Affiliation(s)
| | | | | | | | - Na Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
31
|
Müller KJ, Forbrig R, Reis J, Wiegand L, Barci E, Kunte SC, Kaiser L, Schönecker S, Schichor C, Harter PN, Thon N, von Baumgarten L, Preusser M, Albert NL. Measurable disease as baseline criterion for response assessment in glioblastoma: A comparison of PET -based (PET RANO 1.0) and MRI-based (RANO) assessments. Neuro Oncol 2025; 27:77-88. [PMID: 39561103 PMCID: PMC11726251 DOI: 10.1093/neuonc/noae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Recently, criteria based on amino acid positron emission tomography (PET) have been proposed for response assessment in diffuse gliomas (PET RANO 1.0). In this study, we compare the prevalence of measurable disease according to PET RANO 1.0 with magnetic resonance imaging (MRI)-based Response Assessment in Neuro-Oncology (RANO) criteria in glioblastoma. METHODS We retrospectively identified patients with newly diagnosed IDH-wild-type glioblastoma who underwent [18F] Fluoroethyltyrosine (FET) PET and MRI after resection or biopsy and before radio-/radiochemotherapy. Two independent investigators analyzed measurable disease according to PET RANO 1.0 or MRI-RANO criteria. Additionally, lesion size, congruency patterns, and uptake intensity on [18F]FET PET images were assessed. RESULTS We evaluated 125 patients including 49 cases after primary resection and 76 cases after biopsy. Using PET criteria, 113 out of 125 patients (90.4%) had measurable disease, with a median PET-positive volume of 15.34 cm3 (8.83-38.03). With MRI, a significantly lower proportion of patients had measurable disease (57/125, 45.6%; P < .001) with a median sum of maximum cross-sectional diameters of 35.65 mm (26.18-45.98). None of the 12 patients without measurable disease on PET had measurable disease on MRI. Contrariwise, 56/68 patients (82.4%) without measurable disease on MRI exhibited measurable disease on PET. Clinical performance status correlated significantly with PET-positive volume and MRI-based sum of diameters (P < .0059, P < .0087, respectively). CONCLUSIONS [18F]FET PET identifies a higher number of patients with measurable disease compared to conventional MRI in newly diagnosed glioblastoma. PET-based assessment may serve as a novel baseline parameter for evaluating residual tumor burden and improving patient stratification in glioblastoma studies. Further validation in prospective trials is warranted.
Collapse
Affiliation(s)
- Katharina J Müller
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Forbrig
- Institute of Neuroradiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jonas Reis
- Institute of Neuroradiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lilian Wiegand
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Enio Barci
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sophie C Kunte
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Bayerisches Zentrum für Krebsforschung (BZKF), Partner Site Munich, Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Stephan Schönecker
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christian Schichor
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University/University Hospital, LMU Munich, Munich, Germany
| | - Patrick N Harter
- Bayerisches Zentrum für Krebsforschung (BZKF), Partner Site Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University/University Hospital, LMU Munich, Munich, Germany
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Niklas Thon
- Bayerisches Zentrum für Krebsforschung (BZKF), Partner Site Munich, Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University/University Hospital, LMU Munich, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Bayerisches Zentrum für Krebsforschung (BZKF), Partner Site Munich, Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University/University Hospital, LMU Munich, Munich, Germany
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Bayerisches Zentrum für Krebsforschung (BZKF), Partner Site Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University/University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
32
|
Jelgersma C, Alsolivany J, Akkas G, Wasilewski D, Gastl B, Misch M, Capper D, Kaul D, Bullinger L, Vajkoczy P, Onken J. Real-world experience with TTFields in glioma patients with emphasis on therapy usage. Front Oncol 2025; 14:1430793. [PMID: 39839796 PMCID: PMC11747310 DOI: 10.3389/fonc.2024.1430793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/27/2024] [Indexed: 01/23/2025] Open
Abstract
Tumor Treating Fields (TTFields) has emerged as a significant adjunctive component in the treatment of high-grade gliomas following the EF-14 trial in 2017. The incorporation of TTFields, alongside cyclic temozolomide therapy, has demonstrated improved patient outcomes when the usage exceeds 18 h per day (75% usage). Post-hoc analysis of the EF-14 trial has demonstrated that therapy usage exceeding 90% is associated with an additional benefit, while rates above 50% have also proven effective in literature. Given the cost-intensive nature and mild- to- moderate constraints associated with the therapy, our objective is to generate real-world data on therapy usage through a retrospective analysis at a high-throughput academic center. Between June 2015 and February 2022, a total of 113 high-grade glioma patients received TTFields therapy. Eight patients discontinued TTFields therapy within 2 months with less than 50% usage and were excluded from further analysis. For the remaining patients, the median age was 51 years (range: 20-76 years) and the mean preoperative Karnofsky index was 80%-90%. Most of the patients (75.2%) initiated therapy concurrently with first-line treatment, of whom 27.6% started TTFields therapy concomitant to the first cycle of temozolomide. 15.2% started TTFields therapy in the second-line and 9.5% in the third-line setting. The study cohort had an average therapy duration of 9.3 months with 3.2 break days per month. The mean therapy usage was 65.5% (SD 17.6%). Usage was highest during the first 3 months, with rates of 77.7%, 72.3%, and 71.6%, and then dropped to around 60% in the following 6 months. Linear regression found no predictors of usage, such as age, timing of therapy initiation, and duration or gender. 55% of patients continued TTFields beyond the first recurrence. Interestingly, no drop in usage rates was observed before tumor recurrence was communicated. However, after diagnosis, patients exhibited a significant drop in usage to an average of 52.3%. This high-volume, real-world TTFields usage data reveal that the extent of usage falls short of the intended 75%. It highlights the importance of monitoring and promoting adherence to maximize its potential benefits in managing high-grade glioma patients. Furthermore, strategies to expedite therapy initiation and improve long-term adherence are warranted.
Collapse
Affiliation(s)
- Claudius Jelgersma
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Joan Alsolivany
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Gülsüm Akkas
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - David Wasilewski
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Martin Misch
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - David Capper
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology and Radiotherapy Charité – Universitätsmedizin Berlin, Berlin, Germany
- Radiation Therapy, Health and Medical University Potsdam, Potsdam, Germany
| | - Lars Bullinger
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
33
|
da Silva KC, Lima IS, dos Santos CC, Nonaka CKV, Souza BSDF, David JM, Ulrich H, do Nascimento RP, Costa MDFD, dos Santos BL, Costa SL. Agathisflavone Inhibits Viability and Modulates the Expression of miR-125b, miR-155, IL-6, and Arginase in Glioblastoma Cells and Microglia/Macrophage Activation. Molecules 2025; 30:158. [PMID: 39795214 PMCID: PMC11721753 DOI: 10.3390/molecules30010158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Glioblastomas (GBM) are malignant tumours with poor prognosis. Treatment involves chemotherapy and/or radiotherapy; however, there is currently no standard treatment for recurrence, and prognosis remains unfavourable. Inflammatory mediators and microRNAs (miRNAs) influence the aggressiveness of GBM, being involved in the communication with the cells of the tumour parenchyma, including microglia/macrophages, and maintaining an immunosuppressive microenvironment. Hence, the modulation of miRNAs and inflammatory factors may improve GBM treatments. In this study, we investigated the effects of agathisflavone, a biflavonoid purified from Cenostigma pyramidale (Tul.), on the growth and migration of GBM cells, on the expression of inflammatory cytokines and microRNAs, as well on the response of microglia. Agathisflavone (5-30 μM) induced a dose- and time-dependent reduction in the viability of both human GL-15 and rat C6 cells, as determined by the MTT test, and reduced cell migration, as determined by cell scratch assay. RT-qPCR analysis revealed that agathisflavone (5 μM) down-regulated the expression of miR-125b and miR-155 in the secretome derived from GL-15 cells, which was associated with upregulation of the mRNA expression of IL-6 and arginase-1 immunoregulatory factors. Exposure of human microglia/macrophage to the secretome from GL-15 GMB cells modulated proliferation and morphology, effects that were modulated by agathisflavone treatment. These results demonstrate the effect of flavonoids on the growth of GBM cells, which impacts cells in the microenvironment and can be considered for preclinical studies for adjuvant treatments.
Collapse
Affiliation(s)
- Karina Costa da Silva
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Irlã Santos Lima
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Carolina Kymie Vasques Nonaka
- Center of Biotechnology and Cell Therapy, São Rafael Hospital, D’Or Institute for Research and Teaching, Salvador 41253-190, BA, Brazil; (C.K.V.N.); (B.S.d.F.S.)
| | - Bruno Solano de Freitas Souza
- Center of Biotechnology and Cell Therapy, São Rafael Hospital, D’Or Institute for Research and Teaching, Salvador 41253-190, BA, Brazil; (C.K.V.N.); (B.S.d.F.S.)
- Institute Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, BA, Brazil
| | - Jorge Mauricio David
- Department of General and Inorganic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40231-300, BA, Brazil;
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748-Butantã, São Paulo 05508-900, SP, Brazil;
| | - Ravena Pereira do Nascimento
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-971, RJ, Brazil
| | - Balbino Lino dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
- College of Nursing, Federal University of Vale do São Francisco, Av. José de Sá Maniçoba, S/N, Petrolina 56304-917, PE, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, BA, Brazil; (K.C.d.S.); (I.S.L.); (C.C.d.S.); (R.P.d.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-971, RJ, Brazil
| |
Collapse
|
34
|
Xiao T, Zheng H, Zu K, Yue Y, Wang Y. Tumor-treating fields in cancer therapy: advances of cellular and molecular mechanisms. Clin Transl Oncol 2025; 27:1-14. [PMID: 38884919 DOI: 10.1007/s12094-024-03551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Tumor-Treating Fields (TTFields) use intermediate-frequency and low-intensity electric fields to inhibit tumor cells. However, their mechanisms are still not well understood. This article reviews their key antitumor mechanisms at the cellular and molecular levels, including inhibition of proliferation, induction of death, disturbance of migration, and activation of the immune system. The multifaceted biological effects in combination with other cancer treatments are also summarized. The deep insight into their mechanism will help develop more potential antitumor treatments.
Collapse
Affiliation(s)
- Tong Xiao
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Hao Zheng
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Kaiyang Zu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Youjia Yue
- School of Biomedical Engineeringg, Capital Medical University, Beijing, 100069, China
| | - Ying Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
35
|
Rusak A, Wiatrak B, Krawczyńska K, Górnicki T, Zagórski K, Zadka Ł, Fortuna W. Starting points for the development of new targeted therapies for glioblastoma multiforme. Transl Oncol 2025; 51:102187. [PMID: 39531784 PMCID: PMC11585793 DOI: 10.1016/j.tranon.2024.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive and lethal brain tumors, characterized by rapid growth, invasiveness, and resistance to standard therapies, including surgery, chemotherapy, and radiotherapy. Despite advances in treatment, GBM remains highly resistant due to its complex molecular mechanisms, including angiogenesis, invasion, immune modulation, and lipid metabolism dysregulation. This review explores recent breakthroughs in targeted therapies, focusing on innovative drug carriers such as nanoparticles and liposomes, and their potential to overcome GBM's chemo- and radioresistant phenotypes. We also discuss the molecular pathways involved in GBM progression and the latest therapeutic strategies, including immunotherapy and precision medicine approaches, which hold promise for improving clinical outcomes. The review highlights the importance of understanding GBM's genetic and molecular heterogeneity to develop more effective, personalized treatment protocols aimed at increasing survival rates and enhancing the quality of life for GBM patients.
Collapse
Affiliation(s)
- Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland.
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, J. Mikulicza-Radeckiego 2 Street, Wroclaw 50-345, Poland.
| | - Klaudia Krawczyńska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland.
| | - Tomasz Górnicki
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland
| | - Karol Zagórski
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland
| | - Łukasz Zadka
- Division of Ultrastructural Research, Wroclaw Medical University, T. Chalubinskiego 6a St., Wroclaw 50-368, Poland; Department of Clinical Pharmacology, Wroclaw Medical University, Borowska 211a, Wroclaw 50-556, Poland.
| | - Wojciech Fortuna
- Department of Neurosurgery, Wroclaw Medical University, Borowska 213St, Wroclaw 50-556, Poland.
| |
Collapse
|
36
|
Yamanishi S, Nagashima H, Tanaka K, Uno T, Ikeuchi Y, Iwahashi H, Hashiguchi M, Horii S, Itoh T, Muragaki Y, Sasayama T. Association of preoperative seizures with reduced expression of soluble CD163, an M2 macrophage marker, in the cerebrospinal fluid in isocitrate dehydrogenase wild-type glioblastoma. J Neurooncol 2025; 171:95-103. [PMID: 39377994 DOI: 10.1007/s11060-024-04837-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/19/2024] [Indexed: 01/01/2025]
Abstract
PURPOSE To investigate the relationship between the tumor microenvironment (TME), tumor-related seizures (TRS), and cerebrospinal fluid (CSF) markers that predict preoperative seizures in patients with glioblastoma. METHODS In total, 47 patients with isocitrate dehydrogenase (IDH) wild-type glioblastoma who underwent preoperative CSF examination, 3-T magnetic resonance spectroscopy (MRS), and neurological surgery between January 2017 and December 2023 were included. We measured the concentrations of soluble CD163 (sCD163), a soluble form of the M2 macrophage marker, in the CSF, the metabolite concentration on MRS, and the number of CD163-positive M2 macrophages in the tumor tissue. Factors associated with preoperative seizures were examined. RESULTS Twelve patients (25.5%) had preoperative seizures. sCD163 levels in the CSF were positively correlated with the number of CD163-positive M2 macrophages in the tumor tissue, and both were significantly lower in the preoperative seizure group than in the non-preoperative seizure group (p = 0.0124 and p < 0.0001, respectively). MRS indicated that only glutathione (GSH) concentrations were higher in the preoperative seizure group than in the non-preoperative seizure group (2.55 mM and 1.87 mM, respectively; p = 0.0171). CD163-positive M2 macrophages were inversely correlated with GSH levels. sCD163 in the CSF had a high predictive accuracy (sensitivity, 91.7%; specificity, 54.3%; and area under the receiver operator curve, 0.745) for preoperative seizures. CONCLUSIONS The CSF level of sCD163 is useful for predicting the TME and preoperative seizures in IDH wild-type glioblastoma.
Collapse
Affiliation(s)
- Shunsuke Yamanishi
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hiroaki Nagashima
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takiko Uno
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yusuke Ikeuchi
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hirofumi Iwahashi
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Mitsuru Hashiguchi
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shintaro Horii
- Division of Radiology, Department of Medical Technology, Kobe University Hospital, Kobe, Japan
| | - Tomoo Itoh
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiro Muragaki
- Center for Advanced Medical Engineering Research and Development, Kobe University, Kobe, Japan
| | - Takashi Sasayama
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
37
|
Dhiman A, Rana D, Benival D, Garkhal K. Comprehensive insights into glioblastoma multiforme: drug delivery challenges and multimodal treatment strategies. Ther Deliv 2025; 16:87-115. [PMID: 39445563 PMCID: PMC11703381 DOI: 10.1080/20415990.2024.2415281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and malignant brain tumors, with a high prevalence in elderly population. Most chemotherapeutic agents fail to reach the tumor site due to various challenges. However, smart nanocarriers have demonstrated excellent drug-loading capabilities, enabling them to cross the blood brain tumor barrier for the GBM treatment. Surface modification of nanocarriers has significantly enhanced their potential for targeting therapeutics. Moreover, recent innovations in drug therapies, such as the incorporation of theranostic agents in nanocarriers and antibody-drug conjugates, have offered newer insights for both diagnosis and treatment. This review focuses on recent advances in new therapeutic interventions for GBM, with an emphasis on the nanotheranostics systems to maximize therapeutic and diagnostic outcomes.
Collapse
Affiliation(s)
- Ashish Dhiman
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kalpna Garkhal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| |
Collapse
|
38
|
Yuan B, Kikuchi H. Harnessing Arsenic Derivatives and Natural Agents for Enhanced Glioblastoma Therapy. Cells 2024; 13:2138. [PMID: 39768226 PMCID: PMC11674460 DOI: 10.3390/cells13242138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 01/05/2025] Open
Abstract
Glioblastoma (GBM) is the most common and lethal intracranial tumor in adults. Despite advances in the understanding of the molecular events responsible for disease development and progression, survival rates and mortality statistics for GBM patients have been virtually unchanged for decades and chemotherapeutic drugs used to treat GBM are limited. Arsenic derivatives, known as highly effective anticancer agents for leukemia therapy, has been demonstrated to exhibit cytocidal effects toward GBM cells by inducing cell death, cell cycle arrest, inhibition of migration/invasion, and angiogenesis. Differentiation induction of glioma stem-like cells (GSCs) and inhibition of neurosphere formation have also been attributed to the cytotoxicity of arsenic derivatives. Intriguingly, similar cytotoxic effects against GBM cells and GSCs have also been observed in natural agents such as anthocyanidins, tetrandrine, and bufadienolides. In the current review, we highlight the available data on the molecular mechanisms underlying the multifaceted anticancer activity of arsenic compounds and natural agents against cancer cells, especially focusing on GBM cells and GCSs. We also outline possible strategies for developing anticancer therapy by combining natural agents and arsenic compounds, as well as temozolomide, an alkylating agent used to treat GBM, in terms of improvement of chemotherapy sensitivity and minimization of side effects.
Collapse
Affiliation(s)
- Bo Yuan
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan;
| |
Collapse
|
39
|
Khiabani NA, Doustvandi MA, Story D, Nobari SA, Hajizadeh M, Petersen R, Dunbar G, Rossignol J. Glioblastoma therapy: State of the field and future prospects. Life Sci 2024; 359:123227. [PMID: 39537100 DOI: 10.1016/j.lfs.2024.123227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GB) is a cancerous brain tumor that originates from glial cells and leads to thousands of deaths each year and a five-year survival of only 6.8 %. Treatments for GB include surgery, chemotherapy, radiation, and immunotherapy. GB is an incurable fatal disease, necessitating the development of innovative strategies to find a developing effective therapy. Genetic therapies may be crucial in treating GB by identifying the mutations and amplifications of multiple genes, which drive its proliferation and spread. Use of small interfering RNAs (siRNAs) provides a novel technology used to suppress the genes associated with disease, which forms a basis for targeted therapy in GB and its stem cell population, which are recognized for their ability to develop resistance to chemotherapy and tumorigenic capabilities. This review examines the use of siRNAs in GB, emphasizing their effectiveness in suppressing key oncogenes and signaling pathways associated with tumor development, invasion, stemness, and resistance to standard treatments. siRNA-based gene silencing is a promising approach for developing targeted therapeutics against GB and associated stem cell populations, potentially enhancing patient outcomes and survival rates in this devastating disease.
Collapse
Affiliation(s)
- Nadia Allahyarzadeh Khiabani
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | | | - Darren Story
- Department of Psychology, Saginaw Valley State University, University Center, MI 48710, USA
| | | | | | - Robert Petersen
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Gary Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA.
| |
Collapse
|
40
|
Kumar M, Nassour-Caswell LC, Alrefai H, Anderson JC, Schanel TL, Hicks PH, Cardan R, Willey CD. A High-Throughput Neurosphere-Based Colony Formation Assay to Test Drug and Radiation Sensitivity of Different Patient-Derived Glioblastoma Lines. Cells 2024; 13:1995. [PMID: 39682742 PMCID: PMC11640616 DOI: 10.3390/cells13231995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
The gold standard assay for radiation response is the clonogenic assay, a normalized colony formation assay (CFA) that can capture a broad range of radiation-induced cell death mechanisms. Traditionally, this assay relies on two-dimensional (2D) cell culture conditions with colonies counted by fixing and staining protocols. While some groups have converted these to three-dimensional (3D) conditions, these models still utilize 2D-like media compositions containing serum that are incompatible with stem-like cell models such as brain tumor initiating cells (BTICs) that form self-aggregating spheroids in neural stem cell media. BTICs are the preferred patient-derived model system for studying glioblastoma (GBM) as they tend to better retain molecular and phenotypic characteristics of the original tumor tissue. As such, it is important that preclinical radiation studies should be adapted to BTIC conditions. In this study, we describe a series of experimental approaches for performing CFA experiments with BTIC cultures. Our results indicate that serum-free clonogenic assays are feasible for combination drug and radiation testing and may better facilitate translatability of preclinical findings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christopher D. Willey
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL 35249, USA; (M.K.); (L.C.N.-C.); (H.A.); (J.C.A.); (T.L.S.); (P.H.H.); (R.C.)
| |
Collapse
|
41
|
Guo X, Xing H, Pan H, Wang Y, Chen W, Wang H, Zhang X, Liu J, Xu N, Wang Y, Ma W. Neuronavigation Combined With Intraoperative Ultrasound and Intraoperative Magnetic Resonance Imaging Versus Neuronavigation Alone in Diffuse Glioma Surgery. World Neurosurg 2024; 192:e355-e365. [PMID: 39343380 DOI: 10.1016/j.wneu.2024.09.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVE This study aimed to integrate intraoperative ultrasound and magnetic resonance imaging (IMRI) with neuronavigation (NN) to create a multimodal surgical protocol for diffuse gliomas. Clinical outcomes were compared to the standard NN-guided protocol. METHODS Adult patients with diffuse gliomas scheduled for gross total resection (GTR) were consecutively enrolled to undergo either NN-guided surgery (80 patients, July 2019-January 2022) or multimodal-integrated surgery (80 patients, February 2022-August 2023). The primary outcomes were the extent of resection (EOR) and GTR. Additional outcomes included operative time, blood loss, length of hospital stay, and patient survival. RESULTS GTR was achieved in 69% of patients who underwent multimodal-integrated surgery, compared to 43% of those who received NN-guided surgery (P = 0.002). Residual tumor was detected by IMRI in 53 patients (66%), and further GTR was achieved in 28 of these cases. The median EOR was 100% for the multimodal group and 95% for the NN-guided group (P = 0.001), while the median operative time was 8 hours versus 5 hours (P < 0.001). Neurological deficits, blood loss, and hospital stay durations were comparable between 2 groups. Multimodal-integrated surgery resulted in greater EOR and higher GTR rates in contrast-enhancing gliomas, gliomas in eloquent regions, and large gliomas (≥50 mm). GTR in glioblastomas and other contrast-enhancing gliomas contributed to improved overall survival. CONCLUSIONS Compared to standard NN-guided surgery, multimodal-integrated surgery using NN, IMRI, and intraoperative ultrasound significantly increased the EOR and GTR rates for diffuse gliomas.
Collapse
Affiliation(s)
- Xiaopeng Guo
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; China Anti-Cancer Association Specialty Committee of Glioma, Peking Union Medical College Hospital, Beijing, China
| | - Hao Xing
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huiru Pan
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuekun Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenlin Chen
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Zhang
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiahui Liu
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Xu
- Department of Anesthesia, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; China Anti-Cancer Association Specialty Committee of Glioma, Peking Union Medical College Hospital, Beijing, China.
| | - Wenbin Ma
- Department of Neurosurgery, Center for Malignant Brain Tumors, and National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; China Anti-Cancer Association Specialty Committee of Glioma, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
42
|
Xu Q, Yang C, Wang L, Zhou J. Unveiling the role of RNA methylation in glioma: Mechanisms, prognostic biomarkers, and therapeutic targets. Cell Signal 2024; 124:111380. [PMID: 39236835 DOI: 10.1016/j.cellsig.2024.111380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Gliomas, the most prevalent malignant brain tumors in the central nervous system, are marked by rapid growth, high recurrence rates, and poor prognosis. Glioblastoma (GBM) stands out as the most aggressive subtype, characterized by significant heterogeneity. The etiology of gliomas remains elusive. RNA modifications, particularly reversible methylation, play a crucial role in regulating transcription and translation throughout the RNA lifecycle. Increasing evidence highlights the prevalence of RNA methylation in primary central nervous system malignancies, underscoring its pivotal role in glioma pathogenesis. This review focuses on recent findings regarding changes in RNA methylation expression and their effects on glioma development and progression, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), and N7-methylguanosine (m7G). Given the extensive roles of RNA methylation in gliomas, the potential of RNA methylation-related regulators as prognostic markers and therapeutic targets was also explored, aiming to enhance clinical management and improve patient outcomes.
Collapse
Affiliation(s)
- Qichen Xu
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Zhejiang, China
| | - Chunsong Yang
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Zhejiang, China
| | - Liyun Wang
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Zhejiang, China
| | - Jing Zhou
- Department of Neurosurgery, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Zhejiang, China.
| |
Collapse
|
43
|
Bai F, Deng Y, Li L, Lv M, Razzokov J, Xu Q, Xu Z, Chen Z, Chen G, Chen Z. Advancements and challenges in brain cancer therapeutics. EXPLORATION (BEIJING, CHINA) 2024; 4:20230177. [PMID: 39713205 PMCID: PMC11655316 DOI: 10.1002/exp.20230177] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/02/2024] [Indexed: 12/24/2024]
Abstract
Treating brain tumors requires a nuanced understanding of the brain, a vital and delicate organ. Location, size, tumor type, and surrounding tissue health are crucial in developing treatment plans. This review comprehensively summarizes various treatment options that are available or could be potentially available for brain tumors, including physical therapies (radiotherapy, ablation therapy, photodynamic therapy, tumor-treating field therapy, and cold atmospheric plasma therapy) and non-physical therapies (surgical resection, chemotherapy, targeted therapy, and immunotherapy). Mechanisms of action, potential side effects, indications, and latest developments, as well as their limitations, are highlighted. Furthermore, the requirements for personalized, multi-modal treatment approaches in this rapidly evolving field are discussed, emphasizing the balance between efficacy and patient safety.
Collapse
Affiliation(s)
- Fan Bai
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Advanced Therapeutic CenterNational Innovation Center for Advanced Medical DevicesShenzhenChina
| | - Yueyang Deng
- Department of Biomedical EngineeringMcGill UniversityMontrealQuebecCanada
- Rosalind & Morris Goodman Cancer InstituteMcGill UniversityMontrealQuebecCanada
| | - Long Li
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesShenzhenGuangdongP. R. China
| | - Ming Lv
- Department of Medical EngineeringMedical Supplies Center of Chinese PLA General HospitalBeijingChina
| | - Jamoliddin Razzokov
- Institute of Fundamental and Applied ResearchNational Research University TIIAMETashkentUzbekistan
- Laboratory of Experimental BiophysicsCentre for Advanced TechnologiesTashkentUzbekistan
- Department of Biomedical EngineeringTashkent State Technical UniversityTashkentUzbekistan
| | - Qingnan Xu
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Zhen Xu
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Zhaowei Chen
- Institute of Food Safety and Environment MonitoringMOE Key Laboratory for Analytical Science of Food Safety and BiologyCollege of ChemistryFuzhou UniversityFuzhouChina
| | - Guojun Chen
- Department of Biomedical EngineeringMcGill UniversityMontrealQuebecCanada
- Rosalind & Morris Goodman Cancer InstituteMcGill UniversityMontrealQuebecCanada
| | - Zhitong Chen
- Paul C Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health EngineeringShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Advanced Therapeutic CenterNational Innovation Center for Advanced Medical DevicesShenzhenChina
- University of Chinese Academy of SciencesShenzhenGuangdongP. R. China
- Key Laboratory of Biomedical Imaging Science and SystemChinese Academy of SciencesShenzhenChina
| |
Collapse
|
44
|
Chauhan K, Vijayan V, Pant P, Sharma S, Hassan MI, Ganguly NK, Sharma P, Rana R. Identifying potential inhibitors against galectin-3-binding protein (LGALS3BP) for therapeutic targeting of glioma. J Biomol Struct Dyn 2024:1-13. [PMID: 39589107 DOI: 10.1080/07391102.2024.2431185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/18/2024] [Indexed: 11/27/2024]
Abstract
Glioblastoma is one of the most lethal types of Gliomas, and its treatment greatly depends on the stage of its detection. Galactin-3-binding protein (LGALS3BP) serves as a novel circulatory biomarker for detecting glioma at an early stage. This protein is also responsible for metastasis, proliferative signaling, angiogenesis, and immune system evasion in the case of brain tumors. Inhibition of LGALS3BP can help in the reduction of metastasis and progression of the disease. Currently, no effective drug is available that can completely treat glioma. In this study, we have virtually screened the National Cancer Institute (NCI) drug databank to discover potential inhibitors of LGALS3BP. Based on the binding free energy calculations using MMPBSA, three compounds, 627861 (-16.69 kcal/mol), 329090 (-13.66 kcal/mol), and 627855 (-10.01 kcal/mol), were selected as potent inhibitors. 200 ns MD simulation studies further complemented this study. Finally, we recommend three molecules, 627861, 329090, and 627855, can be potential inhibitors of LGAL3SBP. The structural scaffolds of these molecules can also lead to the optimization of better inhibitors of LGALS3BP and be implicated in the therapeutic management of glioma after desired experimental validations.
Collapse
Affiliation(s)
- Kirti Chauhan
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Viswanathan Vijayan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Pradeep Pant
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
45
|
Kowolik E, Szczygieł D, Szczygieł M, Drzał A, Vemuri K, Olsson AK, Griffioen AW, Nowak-Sliwinska P, Wolnicka-Glubisz A, Elas M. Preclinical Photodynamic Therapy Targeting Blood Vessels with AGuIX ® Theranostic Nanoparticles. Cancers (Basel) 2024; 16:3924. [PMID: 39682113 DOI: 10.3390/cancers16233924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Glioblastoma multiforme (GBM) is the most common highly aggressive, primary malignant brain tumor in adults. Current experimental strategies include photodynamic therapy (PDT) and new drug delivery technologies such as nanoparticles, which could play a key role in the treatment, diagnosis, and imaging of brain tumors. Objectives: The purpose of this study was to test the efficacy of PDT using AGuIX-TPP, a polysiloxane-based nanoparticle (AGuIX) that contains TPP (5,10,15,20-tetraphenyl-21H,23H-porphine), in biological models of glioblastoma multiforme and to investigate the vascular mechanisms of action at multiple complexity levels. Methods: PDT effects were studied in monolayer and spheroid cell culture, as well as tumors in chicken chorioallantoic membranes (CAMs) and in mice were studied. Results: Treatment was effective in both endothelial ECRF and glioma U87 cells, as well as in the inhibition of growth of the glioma spheroids. PDT using AGuIX-TPP inhibited U87 tumors growing in CAM and destroyed their vascularization. The U87 tumors were also grown in nude mice. Their vascular network, as well as oxygen partial pressure, were assessed using ultrasound and EPR oximetry. The treatment damaged tumor vessels and slightly decreased oxygen levels. Conclusions: PDT with AGuIX-TPP was effective against glioma cells, spheroids, and tumors; however, in mice, its efficacy appeared to be strongly related to the presence of blood vessels in the tumor before the treatment.
Collapse
Affiliation(s)
- Ewa Kowolik
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Dariusz Szczygieł
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Małgorzata Szczygieł
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Agnieszka Drzał
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Kalyani Vemuri
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Husargatan 3, SE-75123 Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Husargatan 3, SE-75123 Uppsala, Sweden
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Translational Research Center in Oncohaematology, 1211 Geneva, Switzerland
| | - Agnieszka Wolnicka-Glubisz
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Martyna Elas
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| |
Collapse
|
46
|
Minnaar CA, Szigeti GP, Szasz A. The Synergy of Thermal and Non-Thermal Effects in Hyperthermic Oncology. Cancers (Basel) 2024; 16:3908. [PMID: 39682096 DOI: 10.3390/cancers16233908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Modulated electro-hyperthermia (mEHT) is unique due to its combination of thermal and non-thermal effects. METHOD This report summarizes the literature on the effects of mEHT observed in vitro and in vivo. RESULTS The thermal and electrical heterogeneity of tissues allows the radiofrequency signal to selectively target malignant tissue. The applied modulation appears to activate various apoptotic pathways, predominantly leading to immunogenic cell death (ICD). ICD promotes the release of damage-associated molecular patterns, potentially producing tumour-specific antigen-presenting cells. This abscopal-type effect may target distant metastases while treating the primary tumour locally. This immune memory effect is like vaccination mechanisms. CONCLUSIONS The application of mEHT has the potential to expand from local to systemic disease, enabling the simultaneous treatment of micro- and macro-metastases.
Collapse
Affiliation(s)
- Carrie Anne Minnaar
- Department of Radiation Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Gyula Peter Szigeti
- John von Neumann Faculty of Informatics, Óbuda University, 1034 Budapest, Hungary
- MedTech Innovation and Education Center, University Research and Innovation Center, Óbuda University, 1034 Budapest, Hungary
| | - Andras Szasz
- Department of Biotechnics, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| |
Collapse
|
47
|
Vinogradskiy Y, Bahig H, Bucknell NW, Buchsbaum J, Shu HKG. Conference Report: Review of Clinical Implementation of Advanced Quantitative Imaging Techniques for Personalized Radiotherapy. Tomography 2024; 10:1798-1813. [PMID: 39590941 PMCID: PMC11598114 DOI: 10.3390/tomography10110132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The topic of quantitative imaging in radiation therapy was presented as a "Masterclass" at the 2023 annual meeting of the American Society of Radiation Oncology (ASTRO). Dual-energy computed tomography (CT) and single-positron computed tomography were reviewed in detail as the first portion of the meeting session, with data showing utility in many aspects of radiation oncology including treatment planning and dose response. Positron emission tomography/CT scans evaluating the functional volume of lung tissue so as to provide optimal avoidance of healthy lungs were presented second. Advanced brain imaging was then discussed in the context of different forms of magnetic resonance scanning methods as the third area noted with significant discussion of ongoing research programs. Quantitative image analysis was presented to provide clinical utility for the analysis of patients with head and neck cancer. Finally, quality assurance was reviewed for different forms of quantitative imaging given the critical nature of imaging when numerical valuation, not just relative contrast, plays a crucial role in clinical process and decision-making. Conclusions and thoughts are shared in the conclusion, noting strong data supporting the use of quantitative imaging in radiation therapy going forward and that more studies are needed to move the field forward.
Collapse
Affiliation(s)
- Yevgeniy Vinogradskiy
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Houda Bahig
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Centre Hospitalier de l’Universite de Montreal (CHUM), Montreal, QC H2X 3E4, Canada
| | | | | | - Hui-Kuo George Shu
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 19104, USA
| |
Collapse
|
48
|
Wu Q, Geng Z, Lu J, Wang S, Yu Z, Wang S, Ren X, Guan S, Liu T, Zhu C. Neddylation of protein, a new strategy of protein post-translational modification for targeted treatment of central nervous system diseases. Front Neurosci 2024; 18:1467562. [PMID: 39564524 PMCID: PMC11573765 DOI: 10.3389/fnins.2024.1467562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
Neddylation, a type of protein post-translational modification that links the ubiquitin-like protein NEDD8 to substrate proteins, can be involved in various significant cellular processes and generate multiple biological effects. Currently, the best-characterized substrates of neddylation are the Cullin protein family, which is the core subunit of the Cullin-RING E3 ubiquitin ligase complex and controls many important biological processes by promoting ubiquitination and subsequent degradation of various key regulatory proteins. The normal or abnormal process of protein neddylation in the central nervous system can lead to a series of occurrences of normal functions and the development of diseases, providing an attractive, reasonable, and effective targeted therapeutic strategy. Therefore, this study reviews the phenomenon of neddylation in the central nervous system and summarizes the corresponding substrates. Finally, we provide a detailed description of neddylation involved in CNS diseases and treatment methods that may be used to regulate neddylation for the treatment of related diseases.
Collapse
Affiliation(s)
- Qian Wu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ziang Geng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jun Lu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shisong Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongxue Yu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Siqi Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolin Ren
- Department of Neurosurgery, Shenyang Red Cross Hospital, Shenyang, Liaoning, China
| | - Shu Guan
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tiancong Liu
- Department of Otolaryngology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
49
|
Wen J, Xiong L, Wang S, Qiu X, Cui J, Peng F, Liu X, Lu J, Bian H, Chen D, Chang J, Yao Z, Fan S, Zhou D, Li Z, Liu J, Liu H, Chen X, Chen L. Prediction of intracranial electric field strength and analysis of treatment protocols in tumor electric field therapy targeting gliomas of the brain. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 258:108490. [PMID: 39520874 DOI: 10.1016/j.cmpb.2024.108490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND OBJECTIVE Tumor Electric Field Therapy (TEFT) is a new treatment for glioblastoma cells with significant effect and few side effects. However, it is difficult to directly measure the intracranial electric field generated by TEFT, and the inability to control the electric field intensity distribution in the tumor target area also limits the clinical therapeutic effect of TEFT. It is a safe and effective way to construct an efficient and accurate prediction model of intracranial electric field intensity of TEFT by numerical simulation. METHODS Different from the traditional methods, in this study, the brain tissue was segmented based on the MRI data of patients with retained spatial location information, and the spatial position of the brain tissue was given the corresponding electrical parameters after segmentation. Then, a single geometric model of the head profile with the transducer array is constructed, which is assembled with an electrical parameter matrix containing tissue position information. After applying boundary conditions on the transducer, the intracranial electric field intensity could be solved in the frequency domain. The effects of transducer array mode, load voltage and voltage frequency on the intracranial electric field strength were further analyzed. Finally, planning system software was developed for optimizing TEFT treatment regimens for patients. RESULTS Experimental validation and comparison with existing results demonstrate the proposed method has a more efficient and pervasive modeling approach with higher computational accuracy while preserving the details of MRI brain tissue structure completely. In the optimization analysis of treatment protocols, it was found that increasing the load voltage could effectively increase the electric field intensity in the target area, while the effect of voltage frequency on the electric field intensity was very limited. CONCLUSIONS The results showed that adjusting the transducer array mode was the key method for making targeted treatment plans. The proposed method is capable prediction of intracranial electric field strength with high accuracy and provide guidance for the design of the TEFT therapy process. This study provides a valuable reference for the application of TEFT in clinical practice.
Collapse
Affiliation(s)
- Jun Wen
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Lingzhi Xiong
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Shulu Wang
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Xiaoguang Qiu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Jianqiao Cui
- Hunan Drug Inspection Center, Changsha, Hunan, China
| | - Fan Peng
- Public Course Teaching Department, Changsha Health Vocational College, Changsha 410100, China
| | - Xiang Liu
- Hunan Drug Inspection Center, Changsha, Hunan, China
| | - Jian Lu
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Haikuo Bian
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Jiusheng Chang
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Zhengxi Yao
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Sheng Fan
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Dan Zhou
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Ze Li
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Jialin Liu
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Hongyu Liu
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Xu Chen
- The First Clinical College, China Medical University, Shenyang, China
| | - Ling Chen
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
50
|
Hainfellner A, Borkovec M, Seebrecht L, Neuhauser M, Roetzer-Pejrimovsky T, Greutter L, Surböck B, Hager-Seifert A, Gorka-Vom Hof D, Urbanic-Purkart T, Stultschnig M, Cijan C, Würtz F, Calabek-Wohinz B, Pichler J, Höllmüller I, Leibetseder A, Weis S, Kleindienst W, Seiberl M, Bieler L, Hecker C, Schwartz C, Iglseder S, Heugenhauser J, Nowosielski M, Thomé C, Moser P, Hoffermann M, Loibnegger K, Dieckmann K, Tomschik M, Widhalm G, Rössler K, Marosi C, Wöhrer A, Hainfellner JA, Oberndorfer S. Glioblastoma in the real-world setting: patterns of care and outcome in the Austrian population. J Neurooncol 2024; 170:407-418. [PMID: 39192069 PMCID: PMC11538164 DOI: 10.1007/s11060-024-04808-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
PURPOSE We present results of a retrospective population-based investigation of patterns of care and outcome of glioblastoma patients in Austria. PATIENTS AND METHODS In this nation-wide cooperative project, all Austrian glioblastoma patients newly diagnosed between 2014 and 2018 and registered in the ABTR-SANOnet database were included. Histological typing used criteria of the WHO classification of CNS tumors, 4th edition 2016. Patterns of care were assessed, and all patients were followed until the end of 2019. RESULTS 1,420 adult glioblastoma cases were identified. 813 (57.3%) patients were male and 607 (42.7%) female. Median age at diagnosis was 64 years (range: 18-88). Median overall survival (OS) was 11.6 months in the total cohort and 10.9 months in patients with proven IDH-wildtype. Median OS in the patient group ≤ 65 years receiving postoperative standard of care therapy was 16.1 months. In the patient group > 65 years with postoperative therapy, median OS was 11.2 months. Follow-up ≥ 5 years identified 13/264 (4.9%) long-term survivors. Brain tumor surgery frequently was assisted by 5-aminolevulinic acid (5-ALA) fluorescence (up to 55%). Postoperative treatment was initiated around one month after surgery (median: 31 days) following standardized protocols in 1,041/1,420 (73.3%) cases. In 830 patients (58.5%), concomitant radiochemotherapy was started according to the established standard of care. Treatment in case of progressive disease was considerably variable. 170/1,420 patients (12.0%) underwent a second surgical procedure, 467 (33.0%) received systemic treatment after progression, and 173 (12.2%) were re-irradiated. CONCLUSION Our data illustrate and confirm nation-wide translation of effective standard of care to Austrian glioblastoma patients in the recent past. In the case of progressive disease, highly variable therapeutic approaches were used, most frequently accompanied by anti-angiogenic therapy. Long-term survival was observed in a minor proportion of mostly younger patients who typically had gross total tumor resection, a favorable postoperative ECOG score, and standard of care therapy.
Collapse
Affiliation(s)
- Andreas Hainfellner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria.
| | - Martin Borkovec
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Lukas Seebrecht
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Magdalena Neuhauser
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Thomas Roetzer-Pejrimovsky
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Lisa Greutter
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Birgit Surböck
- Department of Neurology, Clinic Favoriten, Vienna, Austria
| | | | | | | | | | - Clemens Cijan
- Department of Neurology, State Hospital Klagenfurt, Klagenfurt, Austria
| | - Franz Würtz
- Department of Pathology, State Hospital Klagenfurt, Klagenfurt, Austria
| | | | - Josef Pichler
- Department of Internal Medicine and Neuro-Oncology, Neuromed Campus, Kepler University Hospital, Johannes Kepler University of Linz, Linz, Austria
| | - Isolde Höllmüller
- Department of Internal Medicine and Neuro-Oncology, Neuromed Campus, Kepler University Hospital, Johannes Kepler University of Linz, Linz, Austria
| | - Annette Leibetseder
- Department of Neurology, Neuromed Campus, Kepler University Hospital, Johannes Kepler University of Linz, Linz, Austria
| | - Serge Weis
- Division of Neuropathology, Department of Pathology and Molecular Pathology, Neuromed Campus, Kepler University Hospital, and Clinical Research Institute for Neurosciences, Johannes Kepler University of Linz, Linz, Austria
| | - Waltraud Kleindienst
- Department of Neurology, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Michael Seiberl
- Department of Neurology, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Lara Bieler
- Department of Neurology, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Constantin Hecker
- Department of Neurology, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
- Department of Neurosurgery, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Christoph Schwartz
- Department of Neurosurgery, University Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Sarah Iglseder
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Martha Nowosielski
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Moser
- Laboratory of Neuropathology, Tirol Kliniken GmbH, Innsbruck, Austria
| | - Markus Hoffermann
- Department of Neurosurgery, State Hospital Feldkirch, Feldkirch, Austria
| | - Karin Loibnegger
- Department of Radiation Oncology, State Hospital Feldkirch, Feldkirch, Austria
| | - Karin Dieckmann
- Department of Radiation Oncology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Matthias Tomschik
- Department of Neurosurgery, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Georg Widhalm
- Department of Neurosurgery, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Karl Rössler
- Department of Neurosurgery, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Christine Marosi
- Division of Palliative Care, Department of Internal Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Division of Oncology, Department of Internal Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Adelheid Wöhrer
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Johannes A Hainfellner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Medical University Campus AKH 4J, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Stefan Oberndorfer
- Department of Neurology, University Hospital St. Pölten, Dunant-Platz 1, 3100, St. Pölten, Austria.
- Karl Landsteiner Institute for Clinical Neurology and Neuropsychology, Department of Neurology, University Hospital St. Pölten, St. Pölten, Austria.
| |
Collapse
|