1
|
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; 26:398-414. [PMID: 40266436 PMCID: PMC12055647 DOI: 10.1007/s11864-025-01320-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
2
|
Kite T, Yadlapalli V, Herbst J, Karlovits S, Wegner RE, Shepard MJ. Stereotactic radiosurgery for recurrent high-grade gliomas. J Clin Neurosci 2025; 135:111150. [PMID: 40015114 DOI: 10.1016/j.jocn.2025.111150] [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: 01/15/2025] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION High-grade gliomas (WHO Grade III and IV) invariably recur. Standardized management in the recurrent setting is ill defined. Stereotactic radiosurgery (SRS) represents a non-invasive treatment modality. Evidence to date is limited and therefore further evaluation of the role of SRS in recurrent high-grade-gliomas (rHGG) is warranted. METHODS We conducted a retrospective cohort study consisting of 33 patients with rHGGs treated with SRS from January 2020 to June 2024. Baseline demographics, radiosurgical parameters, and outcomes/toxicity data were collected. Descriptive statistics were calculated for all continuous variables. Survival analysis was performed using the Kaplan Meier method. Univariate analysis was performed using Cox proportional hazard model. All statistics were performed in GraphPad Prism (V.10). RESULTS Thirty-three patients with 44 rHGG lesions underwent Gamma Knife SRS with a median of 5 fractions (range:1-5). Overall local control at 3-,6-, and 12-months was 69.9 %, 45.9 %, and 31.9 % respectively. Distant tumor control at 3-,6-, and 12-months was 71.7 %, 48.2 %, and 42.2 %. Global tumor control was at 3-,6, and 12-months was 69.9 %, 45.9 %, 31.9 % respectively. Median OS from the time of SRS was 7 months (95 % CI: 6.65-17.23). Median PFS from the time of SRS was 5.5 months (95 % CI: 4.79-14.31). MGMT methylated status was associated with improved OS (HR: 0.24 95 % CI: 0.07-0.60, P = 0.01). CONCLUSIONS SRS affords reasonable local control in the short term for patients with recurrent HGG who are otherwise poor surgical candidates. Local failure is more common than distant failure, albeit global control is critical in increasing PFS. MGMT methylated status is associated with increased overall survival.
Collapse
Affiliation(s)
- Trent Kite
- Department of Neurosurgery, Allegheny Health Network Neuroscience Institute, Pittsburgh, PA, United States
| | | | - John Herbst
- Division of Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
| | - Stephen Karlovits
- Division of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
| | - Rodney E Wegner
- Division of Radiation Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
| | - Matthew J Shepard
- Department of Neurosurgery, Allegheny Health Network Neuroscience Institute, Pittsburgh, PA, United States.
| |
Collapse
|
3
|
Bien-Möller S, Weidemeier ME, Radke J, Baldauf J, Engeli S, Tzvetkov MV, Schroeder HWS. Case report of three patients with end-stage recurrent glioblastoma treated with meldonium. BJC REPORTS 2025; 3:29. [PMID: 40295665 PMCID: PMC12037855 DOI: 10.1038/s44276-025-00124-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/20/2024] [Accepted: 01/21/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Glioblastoma is the most aggressive primary brain tumor in adults. The prognosis is still very poor with a median survival time less than a year. A growing body of data supports the role for fatty acid oxidation (FAO) in the aggressive behavior of glioblastoma. We have previously shown that meldonium, an orally active compound that impairs FAO, caused significant growth reduction of glioblastoma in mice. Here, we report three cases of experimental meldonium-containing therapy in end-stage recurrent glioblastoma patients. METHODS Three end-stage glioblastoma patients, who had second relapse tumor progression after standard of care therapy, received 500 mg meldonium twice a day on the top of the existing therapy regimen. Tolerability and treatment outcomes were monitored. RESULTS Meldonium was well tolerated by all three patients. One patient experienced long-term growth arrest and maintained clinically stable disease status, currently 24 months into treatment with meldonium. In contrast, the other two patients passed away. CONCLUSIONS The case reports presented here suggest good tolerability and the potential for meldonium to improve outcome in glioblastoma patients. Controlled clinical trials need to follow to evaluate systematically possible benefits from the integration of meldonium into standard glioblastoma treatment protocols.
Collapse
Affiliation(s)
- Sandra Bien-Möller
- Department of General Pharmacology, University Medicine Greifswald, Greifswald, Germany.
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany.
| | - Martin E Weidemeier
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany.
| | - Josefine Radke
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany
| | - Jörg Baldauf
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Engeli
- Department of Clinical Pharmacology, University Medicine Greifswald, Greifswald, Germany
| | - Mladen V Tzvetkov
- Department of General Pharmacology, University Medicine Greifswald, Greifswald, Germany
| | - Henry W S Schroeder
- Department of Neurosurgery, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
4
|
Han W, Chen L. The therapeutic efficacy and application prospects of tumor-treating fields (TTFields) in resolving malignant tumors of central nervous system. Clin Transl Oncol 2025:10.1007/s12094-025-03909-x. [PMID: 40227534 DOI: 10.1007/s12094-025-03909-x] [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: 02/06/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Malignancies in the central nervous system (CNS) are among the most prevalent and lethal tumors. Tumor treating fields (TTFields), a physical therapeutic strategy, show significant potential in treating CNS tumors by inducing cell apoptosis, cell-cycle arrest, immune activation, and enhancing anti-PD-1 therapy efficacy. Additionally, TTFields can increase blood-brain barrier (BBB) permeability, further supporting their application in CNS malignancies. This review aims to summarize the advances and mechanisms of TTFields in CNS tumor treatment while addressing its current limitations and challenges. METHODS We reviewed existing literature on TTFields, focusing on their effects on glioma and brain metastasis (BM)-related primary tumors. The mechanisms investigated included mitosis and cell cycle interference, inhibition of cell migration and invasion, promotion of apoptosis and protective autophagy, activation of immunogenic cell death (ICD) and immune responses, and modulation of BBB permeability. RESULTS TTFields demonstrate inhibitory effects on CNS malignancies, particularly in glioma. They also suppress brain metastasis from primary tumors such as lung cancer, breast cancer, melanoma, and colorectal cancer. Mechanistically, TTFields act through multiple pathways, including disrupting mitosis, impeding cell migration and invasion, enhancing apoptosis and autophagy, activating immune responses, and increasing BBB permeability. CONCLUSION TTFields exhibit therapeutic potential in CNS malignancies, especially glioblastoma (GBM), through diverse biological mechanisms. Their ability to enhance BBB permeability and target metastatic tumors suggests promise for broader clinical applications, including brain metastasis treatment.
Collapse
Affiliation(s)
- Wei Han
- Neurosurgical Department of Huashan Hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
- Tianqiao and Chrissy, Chen Institute Clinical Translational Research Center, Shanghai, 200032, China.
| | - Liang Chen
- Neurosurgical Department of Huashan Hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Tianqiao and Chrissy, Chen Institute Clinical Translational Research Center, Shanghai, 200032, China
| |
Collapse
|
5
|
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
|
6
|
Mason WP, Harrison RA, Lapointe S, Lim-Fat MJ, MacNeil MV, Mathieu D, Perry JR, Pitz MW, Roberge D, Tsang DS, Tsien C, van Landeghem FKH, Zadeh G, Easaw J. Canadian Expert Consensus Recommendations for the Diagnosis and Management of Glioblastoma: Results of a Delphi Study. Curr Oncol 2025; 32:207. [PMID: 40277764 PMCID: PMC12026134 DOI: 10.3390/curroncol32040207] [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: 02/25/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Glioblastoma is the most common and aggressive malignant brain tumor in adults, with an increasing incidence and a poor prognosis. Current challenges in glioblastoma management include rapid tumor growth, limited treatment effectiveness, high recurrence rates, and a significant impact on patients' quality of life. Given the complexity of glioblastoma care and recent advancements in diagnostic and treatment modalities, updated guidelines are needed in Canada. This Delphi study aimed to develop Canadian consensus recommendations for the diagnosis, classification, and management of newly diagnosed and recurrent glioblastoma. A multidisciplinary panel of 14 Canadian experts in glioblastoma care was convened, and a comprehensive literature review was conducted to synthesize evidence and formulate initial recommendations. Consensus was achieved through three Delphi rounds, in which panelists rated their agreement with recommendation statements on a five-point Likert scale. Statements with ≥75% agreement were accepted, and others were revised for re-voting. Final recommendations were formulated based on the consensus level, strength of evidence, clinical expertise, and consideration of the Canadian healthcare context. These recommendations aim to standardize glioblastoma diagnosis and classification across Canada, provide evidence-based guidance for optimal treatment selection, integrate novel therapies, and enhance the overall quality of care for glioblastoma patients.
Collapse
Affiliation(s)
- Warren P. Mason
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Rebecca A. Harrison
- Department of Medicine, University of British Columbia, Vancouver, BC V5Z 4E6, Canada
| | - Sarah Lapointe
- Department of Medicine, Centre Hospitalier Universitaire de Montreal, Montreal, QC H2X 3J4, Canada
- Faculty of Neuroscience, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Mary Jane Lim-Fat
- Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Mary V. MacNeil
- Department of Medicine, Dalhousie University, QE II Health Science Centre, Halifax, NS B3H 2Y9, Canada
- Department of Medicine, Nova Scotia Cancer Care, Halifax, NS B3H 1V8, Canada
| | - David Mathieu
- Department of Surgery, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - James R. Perry
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A4, Canada
| | - Marshall W. Pitz
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - David Roberge
- Division of Radiation Oncology, Centre Hospitalier Universitaire de Montreal, Montreal, QC H2X 0C1, Canada
- Department of Radiology, Radiation-Oncology and Nuclear Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Derek S. Tsang
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Christina Tsien
- Department of Radiation Oncology, McGill University, Montreal, QC H4A 3J1, Canada
| | - Frank K. H. van Landeghem
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Gelareh Zadeh
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Krembil Brain Institute, University Health Network, Toronto, ON M5T 1M8, Canada
| | - Jacob Easaw
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
7
|
Dirven L, Machingura A, van den Bent MJ, Coens C, Bottomley A, Brandes AA, Domont J, Idbaih A, Koekkoek JAF, Reijneveld JC, Platten M, Wick W, Taphoorn MJB. Health-related quality of life in patients with progressive glioblastoma treated with combined bevacizumab and lomustine versus lomustine only: Secondary outcome of the randomized phase III EORTC 26101 study. Neurooncol Pract 2025; 12:209-218. [PMID: 40110057 PMCID: PMC11913650 DOI: 10.1093/nop/npae091] [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] [Indexed: 03/22/2025] Open
Abstract
Background Progression-free survival, but not overall survival, was prolonged with bevacizumab and lomustine compared to lomustine only in the randomized phase 3 European Organization for Research and Treatment of Cancer (EORTC) 26101 study. Objective To evaluate the impact of treatment on health-related quality of life (HRQoL) in progressive glioblastoma patients participating in the EORTC 26101 study. Methods Patients with progressive glioblastoma, after standard radio-chemotherapy, were 2:1 randomized to either BEV/LOM or LOM. HRQoL was a secondary trial outcome and assessed using the EORTC QLQ-C30 and QLQ-BN20 questionnaires at baseline, and subsequently every 12 weeks. Predefined scales for analysis were global health status (GH), physical functioning, social functioning (SF), motor dysfunction, and communication deficit. The primary endpoint was HRQoL during the last assessment up to week 36. Moreover, time to HRQoL deterioration (TTD) and HRQoL deterioration-free survival (DFS) were calculated. Results Out of 437 patients, 402 (92%) patients had a baseline HRQoL assessment, which dropped to 66% at week 36. During the last assessment up to week 36, no differences were observed for predefined scales, apart from SF being clinically relevant lower in the combination arm (mean 66.0 versus 81.0, p = .001). Of note, the baseline SF score was 66.4 for patients in the combination arm, showing stable SF. Median DFS was significantly longer in the combination arm (12.4 weeks) compared to lomustine alone (6.7 weeks), reflecting the difference in time to progression between arms. TTD, not including progression as an event, was not different between treatment arms (median 13.0 versus 12.9 weeks). Conclusion The addition of bevacizumab to lomustine did not negatively affect HRQoL during the progression-free period.
Collapse
Affiliation(s)
- Linda Dirven
- Department of Neurology, Haaglanden Medical Center, The Hague, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Abigirl Machingura
- Quality of Life Department, European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | | | - Corneel Coens
- Quality of Life Department, European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | - Andrew Bottomley
- Quality of Life Department, European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | - Alba A Brandes
- Department of Medical Oncology, AUSL-IRCCS Scienze Neurologiche, Bologna, Italy
| | | | - Ahmed Idbaih
- Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neuro-Oncologie, F-75013, Paris, France
| | - Johan A F Koekkoek
- Department of Neurology, Haaglanden Medical Center, The Hague, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap C Reijneveld
- Department of Neurology and Brain Tumour Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Michael Platten
- UMM, Mannheim, Heidelberg University and CCU Neuroimmunology, Heidelberg, Germany
| | - Wolfgang Wick
- German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neurooncology, German Cancer Research Center, Heidelberg, Germany
- Neurology Clinic and National Centre for Tumour Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin J B Taphoorn
- Department of Neurology, Haaglanden Medical Center, The Hague, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
8
|
Rousseau J, Lapointe S, Roberge D. Tumor-Treating Fields and Related Treatments in the Management of Pediatric Brain Tumors. Curr Oncol 2025; 32:185. [PMID: 40277742 PMCID: PMC12025919 DOI: 10.3390/curroncol32040185] [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: 01/31/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
Pediatric primary brain tumors pose significant therapeutic challenges due to their aggressive nature and the critical environment of the developing brain. Traditional modalities like surgery, chemotherapy, and radiotherapy often achieve limited success in high-grade gliomas and embryonal tumors. Tumor-treating fields (TTfields), a non-invasive therapy delivering alternating electric fields, has emerged as a promising approach to disrupt tumor cell division through mechanisms such as mitotic disruption, DNA damage, and tumor microenvironment modulation. TTfields are thought to selectively target dividing tumor cells while sparing healthy, non-dividing cells. While TTfields therapy is FDA-approved for the management of glioblastoma and other cancers, its application in pediatric brain tumors remains under investigation. Preclinical studies reveal its potential in medulloblastoma and ependymoma models, while observational data suggest its safety and feasibility in children. Current research focuses on optimizing TTfields' efficacy through advanced technologies, including high-intensity arrays, skull remodeling, and integration with immunotherapies such as immune checkpoint inhibitors. Innovative device-based therapies like magnetic field-based technologies further expand the treatment possibilities. As clinical trials progress, TTfields and related modalities offer hope for addressing unmet needs in pediatric neuro-oncology, especially for tumors in challenging locations. Future directions include biomarker identification, tailored protocols, and novel therapeutic combinations to enhance outcomes in pediatric brain tumor management.
Collapse
Affiliation(s)
- Julien Rousseau
- Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet St., Montreal, QC H2X 3E4, Canada
| | | | | |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Damante MA, Vignolles-Jeong J, Finger G, Kreatsoulas D, Cua S, Giglio P, Ong S, Lonser RR, Wu KC, Elder JB. Early Repeat Residual Resection Versus Adjuvant Therapy for Incompletely Resected Glioblastoma: A Case-Control Study. Neurosurgery 2025:00006123-990000000-01517. [PMID: 39982107 DOI: 10.1227/neu.0000000000003381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/19/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Maximal safe resection of newly diagnosed glioblastoma (GBM) optimizes clinical outcomes. For patients who receive biopsy or subtotal resection (STR), early repeat resection (ERR) to improve extent of resection can be considered but is controversial. Oncological outcomes of patients undergoing ERR for residual GBM to patients receiving upfront at least near-total resection were compared. METHODS This case-control study including patients with GBM treated at a single institution identified 3 treatment groups: (1) ERR patients underwent biopsy or STR (<95% cytoreduction), followed by ≥NTR (≥95% cytoreduction) within 8 weeks; (2) control patients underwent upfront ≥NTR; and (3) biopsy with chemoradiation only. ERR and control patients were 1:1 case-control matched by age, performance status, O6-methylguanine-DNA methyltransferase methylation status, isocitrate dehydrogenase mutation status, and completion of standard chemoradiation. Patient demographics, treatments, and oncological outcomes were analyzed. A P-value of <.05 was considered significant. RESULTS A total of 22 ERR patients (16 biopsies and 6 STR) were identified, and each was matched with a control patient. Baseline patient characteristics at presentation did not differ between matched patients. Time from first surgery to chemoradiation was longer in the ERR cohort (1.8 vs 1.1 months, P < .001). Median overall survival (mOS) (17.7 vs 20.3 months, P = .87) and progression-free survival (5.5 vs 4.5 months, P = .25) did not differ between ERR and control groups, respectively. In the biopsy-only group, mOS was 4.1. Univariate Cox-regression analysis suggested age, failure to complete chemoradiation, isocitrate dehydrogenase-wt, and hemorrhage at presentation were independent predictors of mOS, whereas only age and failure to complete chemoradiation remained independent predictors of mOS after multivariate analysis. Surgical complications were similar between cohorts. CONCLUSION ERR achieving ≥95% extent of resection within 8 weeks of initial surgery results in similar oncological outcomes to upfront ≥NTR in a case-control-matched analysis despite delayed initiation in chemoradiation for the ERR cohort.
Collapse
Affiliation(s)
- Mark A Damante
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | | - Guilherme Finger
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Daniel Kreatsoulas
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Santino Cua
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Pierre Giglio
- Division of Neuro-Oncology, Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Shirley Ong
- Division of Neuro-Oncology, Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Russell R Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Kyle C Wu
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - J Bradley Elder
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
13
|
Khagi S, Kotecha R, Gatson NTN, Jeyapalan S, Abdullah HI, Avgeropoulos NG, Batzianouli ET, Giladi M, Lustgarten L, Goldlust SA. Recent advances in Tumor Treating Fields (TTFields) therapy for glioblastoma. Oncologist 2025; 30:oyae227. [PMID: 39401002 PMCID: PMC11883162 DOI: 10.1093/oncolo/oyae227] [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: 03/16/2024] [Accepted: 07/23/2024] [Indexed: 10/15/2024] Open
Abstract
Tumor Treating Fields (TTFields) therapy is a locoregional, anticancer treatment consisting of a noninvasive, portable device that delivers alternating electric fields to tumors through arrays placed on the skin. Based on efficacy and safety data from global pivotal (randomized phase III) clinical studies, TTFields therapy (Optune Gio) is US Food and Drug Administration-approved for newly diagnosed (nd) and recurrent glioblastoma (GBM) and Conformité Européenne-marked for grade 4 glioma. Here we review data on the multimodal TTFields mechanism of action that includes disruption of cancer cell mitosis, inhibition of DNA replication and damage response, interference with cell motility, and enhancement of systemic antitumor immunity (adaptive immunity). We describe new data showing that TTFields therapy has efficacy in a broad range of patients, with a tolerable safety profile extending to high-risk subpopulations. New analyses of clinical study data also confirmed that overall and progression-free survival positively correlated with increased usage of the device and dose of TTFields at the tumor site. Additionally, pilot/early phase clinical studies evaluating TTFields therapy in ndGBM concomitant with immunotherapy as well as radiotherapy have shown promise, and new pivotal studies will explore TTFields therapy in these settings. Finally, we review recent and ongoing studies in patients in pediatric care, other central nervous system tumors and brain metastases, as well as other advanced-stage solid tumors (ie, lung, ovarian, pancreatic, gastric, and hepatic cancers), that highlight the broad potential of TTFields therapy as an adjuvant treatment in oncology.
Collapse
Affiliation(s)
- Simon Khagi
- Hoag Family Cancer Institute, Newport Beach, CA, United States
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, United States
| | - Na Tosha N Gatson
- Neuro-Oncology Center of Excellence, Indiana University School of Medicine, Indianapolis, IN, United States
- IU Health Neuroscience & Simon Cancer Institutes, Indianapolis, IN, United States
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | | | | | | | | | | | | | - Samuel A Goldlust
- Department of Neuro-Oncology, Saint Luke’s Cancer Institute, Kansas City, MO, United States
| |
Collapse
|
14
|
Hu X, Qiu Z, Yang Y, Xu T, Sheng K, Lu W, Xie J, Xu B. Implantable Ultrasound-Powered MXene/PVA Hydrogel-Based Generator for Treatment of Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2309610. [PMID: 39665226 PMCID: PMC11791951 DOI: 10.1002/advs.202309610] [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: 12/11/2023] [Revised: 06/17/2024] [Indexed: 12/13/2024]
Abstract
Glioblastoma (GBM) is a lethal disease with a poor prognosis due to its strong infiltration, which makes it difficult to remove completely. In this study, an implantable, modulus-tunable, and ultrasound-powered MXene/PVA hydrogel-based tumor treatment device (UP-MPH-TTD), which generates specific electromagnetic alternating fields that disrupt the mitosis of cancer cells without adversely affecting normal neurons is developed. The MXene/PVA hydrogel is used to form a tumor treatment field due to its high biocompatibility, excellent flexibility, and high conductivity, which improves ultrasonic electrical conversion efficiency and significantly reduces the size of the equipment. The implantable UP-MPH-TTD is wirelessly ultrasound-powered, small-sized, lightweight, and simply structured, significantly boosting therapeutic efficiency and reducing restrictions on patient movement. In vitro and in vivo experiments confirmed the device's therapeutic effect, demonstrating a ≈92% inhibition rate in the growth of clinical tumor cells and a 73% reduction in tumor area in tumor-bearing mice. The promising results indicate the broad application potential of the device in the treatment and prognostic improvement of GBM.
Collapse
Affiliation(s)
- Xiaoping Hu
- School of Biomedical EngineeringSun Yat‐sen UniversityNo. 135, Xingang Xi RoadGuangzhou510275P. R. China
- School of Biomedical EngineeringShenzhen Campus of Sun Yat‐sen UniversityNo.66, Gongchang Road, Guangming DistrictShenzhen518107P. R. China
| | - Ziyi Qiu
- School of Biomedical EngineeringSun Yat‐sen UniversityNo. 135, Xingang Xi RoadGuangzhou510275P. R. China
- School of Biomedical EngineeringShenzhen Campus of Sun Yat‐sen UniversityNo.66, Gongchang Road, Guangming DistrictShenzhen518107P. R. China
| | - Yilin Yang
- Faculty of EngineeringDepartment of Electrical and Electronic EngineeringThe University of Hong KongPokfulamHong Kong SAR999077P. R. China
| | - Ting Xu
- School of Biomedical EngineeringShenzhen Campus of Sun Yat‐sen UniversityNo.66, Gongchang Road, Guangming DistrictShenzhen518107P. R. China
| | - Kai Sheng
- School of Biomedical EngineeringSun Yat‐sen UniversityNo. 135, Xingang Xi RoadGuangzhou510275P. R. China
- School of Biomedical EngineeringShenzhen Campus of Sun Yat‐sen UniversityNo.66, Gongchang Road, Guangming DistrictShenzhen518107P. R. China
| | - Weicheng Lu
- Department of AnesthesiologyState Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
| | - Jingdun Xie
- Department of AnesthesiologyState Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouGuangdong510060P. R. China
| | - Bingzhe Xu
- School of Biomedical EngineeringSun Yat‐sen UniversityNo. 135, Xingang Xi RoadGuangzhou510275P. R. China
- School of Biomedical EngineeringShenzhen Campus of Sun Yat‐sen UniversityNo.66, Gongchang Road, Guangming DistrictShenzhen518107P. R. China
| |
Collapse
|
15
|
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
|
16
|
Su H, Peng Y, Wu Y, Zeng X. Overcoming immune evasion with innovative multi-target approaches for glioblastoma. Front Immunol 2025; 16:1541467. [PMID: 39911397 PMCID: PMC11794508 DOI: 10.3389/fimmu.2025.1541467] [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: 12/07/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Glioblastoma (GBM) cells leverage complex endogenous and environmental regulatory mechanisms to drive proliferation, invasion, and metastasis. Tumor immune evasion, facilitated by a multifactorial network, poses a significant challenge to effective therapy, as evidenced by the limited clinical benefits of monotherapies, highlighting the adaptive nature of immune evasion. This review explores glioblastoma's immune evasion mechanisms, the role of ICIs in the tumor microenvironment, and recent clinical advancements, offering theoretical insights and directions for monotherapy and combination therapy in glioblastoma management.
Collapse
Affiliation(s)
- Hai Su
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Peng
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yilong Wu
- Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoli Zeng
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi “Flagship” Oncology Department of Synergy for Chinese and Western Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Oncology, Jiangxi Clinical Research Center for Cancer, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Tanzhu G, Chen L, Ning J, Xue W, Wang C, Xiao G, Yang J, Zhou R. Metastatic brain tumors: from development to cutting-edge treatment. MedComm (Beijing) 2025; 6:e70020. [PMID: 39712454 PMCID: PMC11661909 DOI: 10.1002/mco2.70020] [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: 06/27/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 12/24/2024] Open
Abstract
Metastatic brain tumors, also called brain metastasis (BM), represent a challenging complication of advanced tumors. Tumors that commonly metastasize to the brain include lung cancer and breast cancer. In recent years, the prognosis for BM patients has improved, and significant advancements have been made in both clinical and preclinical research. This review focuses on BM originating from lung cancer and breast cancer. We briefly overview the history and epidemiology of BM, as well as the current diagnostic and treatment paradigms. Additionally, we summarize multiomics evidence on the mechanisms of tumor occurrence and development in the era of artificial intelligence and discuss the role of the tumor microenvironment. Preclinically, we introduce the establishment of BM models, detailed molecular mechanisms, and cutting-edge treatment methods. BM is primarily treated with a comprehensive approach, including local treatments such as surgery and radiotherapy. For lung cancer, targeted therapy and immunotherapy have shown efficacy, while in breast cancer, monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates are effective in BM. Multiomics approaches assist in clinical diagnosis and treatment, revealing the complex mechanisms of BM. Moreover, preclinical agents often need to cross the blood-brain barrier to achieve high intracranial concentrations, including small-molecule inhibitors, nanoparticles, and peptide drugs. Addressing BM is imperative.
Collapse
Affiliation(s)
- Guilong Tanzhu
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Liu Chen
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jiaoyang Ning
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Wenxiang Xue
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunJilinChina
| | - Ce Wang
- Department of RadiologyChina‐Japan Friendship HospitalBeijingChina
| | - Gang Xiao
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jie Yang
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Department of DermatologyXiangya HospitalCentral South UniversityChangshaChina
| | - Rongrong Zhou
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Xiangya Lung Cancer CenterXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan ProvinceChina
| |
Collapse
|
20
|
Kinslow CJ, Mehta MP. Future Directions in the Treatment of Low-Grade Gliomas. Cancer J 2025; 31:e0759. [PMID: 39841425 DOI: 10.1097/ppo.0000000000000759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
ABSTRACT There is major interest in deintensifying therapy for isocitrate dehydrogenase-mutant low-grade gliomas, including with single-agent cytostatic isocitrate dehydrogenase inhibitors. These efforts need head-to-head comparisons with proven modalities, such as chemoradiotherapy. Ongoing clinical trials now group tumors by intrinsic molecular subtype, rather than classic clinical risk factors. Advances in imaging, surgery, and radiotherapy have improved outcomes in low-grade gliomas. Emerging biomarkers, targeted therapies, immunotherapy, radionuclides, and novel medical devices are a promising frontier for future treatment. Diverse representation in glioma research and clinical trials will help to ensure that advancements in care are realized by all groups.
Collapse
Affiliation(s)
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| |
Collapse
|
21
|
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
|
22
|
Schettini F, Pineda E, Rocca A, Buché V, Donofrio CA, Mazariegos M, Ferrari B, Tancredi R, Panni S, Cominetti M, Di Somma A, González J, Fioravanti A, Venturini S, Generali D. Identifying the best treatment choice for relapsing/refractory glioblastoma: a systematic review with multiple Bayesian network meta-analyses. Oncologist 2024:oyae338. [PMID: 39674575 DOI: 10.1093/oncolo/oyae338] [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: 02/23/2024] [Accepted: 11/11/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Glioblastoma is a highly aggressive primary central nervous system tumor characterized by poor outcomes. In case of relapse or progression to adjuvant chemotherapy, there is no univocal preferred regimen for relapsing glioblastoma. METHODS We conducted a systematic review and Bayesian trial-level network meta-analyses (NMA) to identify the regimens associated with the best outcomes. The primary endpoint was overall survival (OS). Secondary endpoints were progression-free survival (PFS) and overall response rates (ORR). We estimated separate treatment rankings based on the surface under the cumulative ranking curve values. Only phase II/III prospective comparative trials were included. RESULTS Twenty-four studies (3733 patients and 27 different therapies) were ultimately included. Twenty-three different regimens were compared for OS, 21 for PFS, and 26 for ORR. When taking lomustine as a common comparator, only regorafenib was likely to be significantly superior in terms of OS (hazard ratio: 0.50, 95% credible interval: 0.33-0.75). Regorafenib was significantly superior to other 16 (69.6%) regimens, including NovoTTF-100A, bevacizumab monotherapy, and several bevacizumab-based combinations. Regarding PFS and ORR, no treatment was clearly superior to the others. CONCLUSIONS This NMA supports regorafenib as one of the best available options for relapsing/refractory glioblastoma. Lomustine, NovoTTF-100A, and bevacizumab emerge as other viable alternative regimens. However, evidence on regorafenib is controversial at best. Moreover, most studies were underpowered, with varying inclusion criteria and primary endpoints, and no longer adapted to the most recent glioblastoma classification. A paradigmatic change in clinical trials' design for relapsing/refractory glioblastoma and more effective treatments are urgently required.
Collapse
Affiliation(s)
- Francesco Schettini
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors Group, Clinic Barcelona Research Foundation-August Pi i Sunyer Biomedical Research Institute (FRCB-IDIBAPS), 08036, Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, 08036, Spain
| | - Estela Pineda
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors Group, Clinic Barcelona Research Foundation-August Pi i Sunyer Biomedical Research Institute (FRCB-IDIBAPS), 08036, Barcelona, Spain
| | - Andrea Rocca
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147 Trieste, Italy
- Hospital of Cattinara, University of Trieste, Trieste, 34149, Italy
| | - Victoria Buché
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147 Trieste, Italy
| | - Carmine Antonio Donofrio
- Neurosurgery, ASST Cremona, 26100 Cremona, Italy
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Manuel Mazariegos
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036, Barcelona, Spain
| | | | | | - Stefano Panni
- Breast and Brain Unit, ASST Cremona, 26100 Cremona, Italy
| | | | - Alberto Di Somma
- Department of Neurosurgery, Hospital Clinic of Barcelona, Barcelona, 08036, Spain
| | - Josep González
- Department of Neurosurgery, Hospital Clinic of Barcelona, Barcelona, 08036, Spain
| | | | - Sergio Venturini
- Department of Economic and Social Sciences, Catholic University of Sacred Heart - Cremona Campus, 26100 Cremona, Italy
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147 Trieste, Italy
- Breast and Brain Unit, ASST Cremona, 26100 Cremona, Italy
| |
Collapse
|
23
|
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
|
24
|
Sheppard DP, Noll KR, Wefel JS, Bradshaw ME. Neuropsychological Evaluation for Oncology. Neurol Clin 2024; 42:875-887. [PMID: 39343481 PMCID: PMC11443061 DOI: 10.1016/j.ncl.2024.05.012] [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: 10/01/2024]
Abstract
Cognitive dysfunction is common in cancers and their treatments. Factors that can contribute to cognitive dysfunction include direct and indirect effects of cancer, surgery, radiation, systemic therapy, as well as comorbidities, fatigue, and mood disturbance. Using objective, validated measures, a neuropsychological evaluation can provide information regarding patterns of cognitive function. Emphasis of cognitive domains assessed may vary depending on disease and treatment history. Cognitive interventions can minimize the effects of cancer-related cognitive dysfunction on daily life.
Collapse
Affiliation(s)
- David P Sheppard
- Department of Rehabilitation Medicine, University of Washington, 1959 Northeast Pacific Street Box 356490, Seattle, WA 98195, USA
| | - Kyle R Noll
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX 77030, USA
| | - Jeffrey S Wefel
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX 77030, USA
| | - Mariana E Bradshaw
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Cai Z, Yang Z, Wang Y, Li Y, Zhao H, Zhao H, Yang X, Wang C, Meng T, Tong X, Zheng H, He Z, Niu C, Yang J, Chen F, Yang Z, Zou Z, Li W. Tumor treating induced fields: a new treatment option for patients with glioblastoma. Front Neurol 2024; 15:1413236. [PMID: 39484048 PMCID: PMC11524832 DOI: 10.3389/fneur.2024.1413236] [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: 04/06/2024] [Accepted: 09/10/2024] [Indexed: 11/03/2024] Open
Abstract
Purpose Currently, a range of electromagnetic therapies, including magnetic field therapy, micro-currents therapy, and tumor treating fields, are under investigation for their potential in central nervous system tumor research. Each of these electromagnetic therapies possesses distinct effects and limitations. Our focus is on overcoming these limitations by developing a novel electric field generator. This generator operates by producing alternating induced currents within the tumor area through electromagnetic induction. Methods Finite element analysis was employed to calculate the distribution of electric fields. Cell viability was assessed using the CCK-8 assay. Tumor volumes and weights served as indicators to evaluate the effectiveness of TTIF. The in-vivo imaging system was utilized to confirm tumor growth in the brains of mice. Results TTIF significantly inhibited the proliferation of U87 cells both in vitro and in vivo. Conclusion TTIF significantly inhibited the proliferation of U87 cells both in vitro and in vivo. Consequently, TTIF emerges as a potential treatment option for patients with progressive or metastatic GBM.
Collapse
Affiliation(s)
- Zehao Cai
- Department of Neuro-oncology Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zukai Yang
- School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Ying Wang
- School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Ye Li
- Kunlun Tripot (Beijing) Medical Technology Co., Ltd., Beijing, China
| | - Hong Zhao
- Kunlun Tripot (Beijing) Medical Technology Co., Ltd., Beijing, China
| | - Hanwen Zhao
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xue Yang
- Department of Neuro-oncology Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Can Wang
- Department of Neuro-oncology Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tengteng Meng
- Kunlun Tripot (Beijing) Medical Technology Co., Ltd., Beijing, China
| | - Xiao Tong
- School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Hao Zheng
- School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Zhaoyong He
- School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Chunli Niu
- Kunlun Tripot (Beijing) Medical Technology Co., Ltd., Beijing, China
| | - Junzhi Yang
- Kunlun Tripot (Beijing) Medical Technology Co., Ltd., Beijing, China
| | - Feng Chen
- Department of Neuro-oncology Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhi Yang
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Zhige Zou
- School of Integrated Circuits, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenbin Li
- Department of Neuro-oncology Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Rodriguez SMB, Tataranu LG, Kamel A, Turliuc S, Rizea RE, Dricu A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int J Mol Sci 2024; 25:10765. [PMID: 39409094 PMCID: PMC11477435 DOI: 10.3390/ijms251910765] [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: 08/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma is known to be one of the most aggressive and fatal human cancers, with a poor prognosis and resistance to standard treatments. In the last few years, many solid tumor treatments have been revolutionized with the help of immunotherapy. However, this type of treatment has failed to improve the results in glioblastoma patients. Effective immunotherapeutic strategies may be developed after understanding how glioblastoma achieves tumor-mediated immune suppression in both local and systemic landscapes. Biomarkers may help identify patients most likely to benefit from this type of treatment. In this review, we discuss the use of immunotherapy in glioblastoma, with an emphasis on immune checkpoint inhibitors and the factors that influence clinical response. A Pubmed data search was performed for all existing information regarding immune checkpoint inhibitors used for the treatment of glioblastoma. All data evaluating the ongoing clinical trials involving the use of ICIs either as monotherapy or in combination with other drugs was compiled and analyzed.
Collapse
Affiliation(s)
- Silvia Mara Baez Rodriguez
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Amira Kamel
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy “G. T. Popa”, 700115 Iasi, Romania;
| | - Radu Eugen Rizea
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania;
| |
Collapse
|
27
|
Motomura K, Sasaki K, Sugii N, Yamaguchi S, Inoue H, Oshima A, Tanaka K, Otani Y, Shirahata M, Shibahara I, Nagane M, Tsuzuki S, Matsutani T, Tsukamoto Y, Kijima N, Asano K, Ohno M, Inoue A, Mineharu Y, Miyake K, Mitobe Y, Hanihara M, Kawanishi Y, Deguchi S, Saito M, Matsuda R, Ujifuku K, Arita H, Sato Y, Yamashita S, Yonezawa U, Yamaguchi J, Momii Y, Ogawa T, Kambe A, Ohba S, Fukai J, Saito N, Kinoshita M, Sumi K, Otani R, Uzuka T, Takebe N, Koizumi S, Saito R, Arakawa Y, Narita Y, the Members of Japan Clinical Oncology Group Brain Tumor Study Group (JCOG-BTSG). Cost of medical care for malignant brain tumors at hospitals in the Japan Clinical Oncology Group brain-tumor study group. Jpn J Clin Oncol 2024; 54:1123-1131. [PMID: 39223700 PMCID: PMC11456849 DOI: 10.1093/jjco/hyae116] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND This study aimed to investigate what treatment are selected for malignant brain tumors, particularly glioblastoma (GBM) and primary central nervous system lymphoma (PCNSL), in real-world Japan and the costs involved. METHODS We conducted a questionnaire survey regarding treatment selections for newly diagnosed GBM and PCNSL treated between July 2021 and June 2022 among 47 institutions in the Japan Clinical Oncology Group-Brain Tumor Study Group. We calculated the total cost and cost per month of the initial therapy for newly diagnosed GBM or PCNSL. RESULTS The most used regimen (46.8%) for GBM in patients aged ≤74 years was 'Surgery + radiotherapy concomitant with temozolomide'. This regimen's total cost was 7.50 million JPY (Japanese yen). Adding carmustine wafer implantation (used in 15.0%), TTFields (used in 14.1%), and bevacizumab (BEV) (used in 14.5%) to the standard treatment of GBM increased the cost by 1.24 million JPY for initial treatment, and 1.44 and 0.22 million JPY per month, respectively. Regarding PCNSL, 'Surgery (biopsy) + rituximab, methotrexate, procarbazine, and vincristine (R-MPV) therapy' was the most used regimen (42.5%) for patients of all ages. This regimen incurred 1.07 million JPY per month. The three PCNSL regimens based on R-MPV therapy were in ultra-high-cost medical care (exceeding 1 million JPY per month). CONCLUSIONS Treatment of malignant brain tumors is generally expensive, and cost-ineffective treatments such as BEV are frequently used. We believe that the results of this study can be used to design future economic health studies examining the cost-effectiveness of malignant brain tumors.
Collapse
Affiliation(s)
- Kazuya Motomura
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Keita Sasaki
- JCOG Data Center/Operations Office, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Narushi Sugii
- Department of Neurosurgery, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Japan
| | - Shigeru Yamaguchi
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, Japan
| | - Hirotaka Inoue
- Department of Neurosurgery, Graduate School of Medical Sciences, 1-1-1, Honjo, Chuo-ku, Kumamoto University, Kumamoto, Japan
| | - Akito Oshima
- Department of Neurosurgery, Yokohama City University, 3-9, Fukuura, Kanazawa, Yokohama, Japan
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Hospital, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, kita-ku, Okayama, Japan
| | - Mitsuaki Shirahata
- Department of Neurosurgery/Neuro-oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, Japan
| | - Ichiyo Shibahara
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara, Japan
| | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, Japan
| | - Shunsuke Tsuzuki
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Japan
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Japan
| | - Noriyuki Kijima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Kenichiro Asano
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, Zaifu-cho 5, Hirosaki, Japan
| | - Makoto Ohno
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Akihiro Inoue
- Department of Neurosurgery, Ehime university school of medicine, 454, Shitsukawa, Toon, Ehime, Japan
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, Japan
| | - Keisuke Miyake
- Department of Neurological Surgery, Kagawa University Faculty of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, Japan
| | - Yuta Mitobe
- Department of Neurosurgery, Yamagata University Hospital, 2-2-2, Iida-Nishi, Yamagata, Japan
| | - Mitsuto Hanihara
- Department of Neurosurgery, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Yu Kawanishi
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, Japan
| | - Shoichi Deguchi
- Department of Neurosurgery, Shizuoka Cancer Center, Shimonagakubo 1007, Nagaizumi, Sunto-gun, Shizuoka, Japan
| | - Masato Saito
- Department of Neurosurgery, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, Japan
| | - Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Shijo-cho 840, Kashihara, Nara, Japan
| | - Kenta Ujifuku
- Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Hideyuki Arita
- Department of Neurosurgery, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka, Japan
| | - Yuichi Sato
- Department of Neurosurgery, Iwate Medical University School of Medicine, 2-1-1 Idai-dori, Yahaba, Shiwa, Iwate, Japan
| | - Shinji Yamashita
- Department of Neurosurgery, Miyazaki University, 5200 Kihara Kiyotake, Miyazaki, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Hiroshima University Hospital, 1-2-3 Minamiku Kasumi, Hiroshima, Japan
| | - Junya Yamaguchi
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Yasutomo Momii
- Department of Neurosurgery, Oita University Faculty of Medicine, 1-1 Idaigaoka Hasama-machi Yufu, Oita, Japan
| | - Takahiro Ogawa
- Department of Neurosurgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Atsushi Kambe
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago, Tottori, Japan
| | - Shigeo Ohba
- Department of Neurosurgery, Fujita Health University, 1-98Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, Japan
| | - Junya Fukai
- Department of Neurological Surgery, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, Japan
| | - Norihiko Saito
- Department of Neurosurgery, Toho University Ohashi Medical Center, 2-22-36, Ohashi, Meguro, Tokyo, Japan
| | - Masashi Kinoshita
- Department of Neurosurgery, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Koichiro Sumi
- Department of Neurological Surgery, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Ryohei Otani
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, Japan
| | - Takeo Uzuka
- Department of Neurosurgery, Dokkyo Medical University Hospital, 880 Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi, Japan
| | - Noriyoshi Takebe
- Department of Neurosurgery, Medical Research Institute Kitano Hospital, PIIF Tazuke-kofukai, 2-4-20 Ohgimachi, Kita-ku, Osaka, Japan
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka, Japan
| | - Ryuta Saito
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, Japan
| | | |
Collapse
|
28
|
Yang X, Hu C, Li L. Technical note: Computational study on thermal management schemes for tumor-treating fields therapy. Med Phys 2024; 51:7632-7644. [PMID: 39023183 DOI: 10.1002/mp.17296] [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: 12/08/2023] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND The study focuses on thermal management in tumor-treating fields (TTFields) therapy, crucial for patient compliance and therapeutic effectiveness. TTFields therapy, an established treatment for glioblastoma, involves applying alternating electric fields to the brain. However, managing the thermal effects generated by electrodes is a major challenge, impacting patient comfort and treatment efficiency. PURPOSE This research aims to explore methods for controlling temperature increases during TTFields therapy without reducing its duty cycle. The study emphasizes optimizing electrode configurations and array arrangements to mitigate temperature rise, thereby maintaining therapy effectiveness and patient compliance. METHODS Using a simplified multi-layer tissue model and finite element analysis, various electrode configurations and array shapes were tested in COMSOL Multiphysics v6.0. Adjustments included changing the electrode gel layer radius from 8 to 12 mm, electrode spacing, and transitioning to a more uniform array arrangement, such as a square array or a circular array. RESULTS The study revealed a strong correlation between high temperatures and edge current density distributions on electrodes. It was found that increasing the electrode gel layer's diameter, enlarging electrode spacing, and adopting a uniform array arrangement markedly mitigated temperature rises. By increasing the gel layer radius from the original 10 to 12 mm, a reduction in the peak temperature increases of approximately 0.3°C was observed. Changing the layout from rectangular to circular with the same area further reduced the peak temperature rise by 0.5°C. Additionally, enlarging the spacing between electrodes also contributed to temperature control. By integrating these strategies, we designed a new circular electrode array with an electrode spacing of 45 mm and a gel radius of 12 mm, successfully reducing the peak temperature from 42.1°C to 40.8°C, effectively achieving temperature control. CONCLUSIONS The research demonstrates that improving electrode and array configurations can effectively manage temperature in TTFields therapy without compromising treatment duration. This strategy is crucial as TTFields therapy relies on prolonged field exposure for effectiveness. The findings offer valuable insights into thermal management in electrode array design and could lead to enhanced patient compliance and treatment efficacy in TTFields therapy.
Collapse
Affiliation(s)
- Xin Yang
- National Engineering Research Centre of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Chunhua Hu
- National Engineering Research Centre of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Luming Li
- National Engineering Research Centre of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
- IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China
- Changping Laboratory, Beijing, China
| |
Collapse
|
29
|
Lerner A, Palmer K, Campion T, Millner TO, Scott E, Lorimer C, Paraskevopoulos D, McKenna G, Marino S, Lewis R, Plowman N. Gliomas in adults: Guidance on investigations, diagnosis, treatment and surveillance. Clin Med (Lond) 2024; 24:100240. [PMID: 39233205 PMCID: PMC11418107 DOI: 10.1016/j.clinme.2024.100240] [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: 06/20/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 09/06/2024]
Abstract
Primary brain tumours are rare but carry a significant morbidity and mortality burden. Malignant gliomas are the most common subtype and their incidence is increasing within our ageing population. The diagnosis and treatment of gliomas involves substantial interplay between multiple specialties, including general medical physicians, radiologists, pathologists, surgeons, oncologists and allied health professionals. At any point along this pathway, patients can present to acute medicine with complications of their cancer or anti-cancer therapy. Increasing the awareness of malignant gliomas among general physicians is paramount to delivering prompt radiological and histopathological diagnoses, facilitating access to earlier and individualised treatment options and allows for effective recognition and management of anticipated complications. This article discusses evidence-based real-world practice for malignant gliomas, encompassing patient presentation, diagnostic pathways, treatments and their complications, and prognosis to guide management outside of specialist centres.
Collapse
Affiliation(s)
| | | | - Tom Campion
- Imaging Department, Barts Health NHS Trust, United Kingdom
| | - Thomas O Millner
- Blizard Institute, Queen Mary University of London and Barts Health NHS Trust, United Kingdom
| | | | | | | | | | - Silvia Marino
- Blizard Institute, Queen Mary University of London and Barts Health NHS Trust, United Kingdom
| | | | | |
Collapse
|
30
|
Goldman MJ, Baskin AM, Sharpe MA, Baskin DS. Advances in gene therapy for high-grade glioma: a review of the clinical evidence. Expert Rev Neurother 2024; 24:879-895. [PMID: 39090786 DOI: 10.1080/14737175.2024.2376847] [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: 03/05/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION High-grade glioma (HGG) is one of the most deadly and difficult cancers to treat. Despite intense research efforts, there has not been a significant breakthrough in treatment outcomes since the early 2000's. Anti-glioma gene therapy has demonstrated promise in preclinical studies and is under investigation in numerous clinical trials. AREAS COVERED This manuscript reviews the current landscape of clinical trials exploring gene therapy treatment of HGG. Using information from clinicaltrials.gov, all trials initiated within the past 5 years (2018-2023) as well as other important trials were cataloged and reviewed. This review discusses trial details, innovative methodologies, and concurrent pharmacological interventions. The review also delves into the subtypes of gene therapy used, trends over time, and future directions. EXPERT OPINION Trials are in the early stages (phase I or II), and there are reports of clinical efficacy in published results. Synergistic effects utilizing immunotherapy within or alongside gene therapy are emerging as a promising avenue for future breakthroughs. Considerable heterogeneity exists across trials concerning administration route, vector selection, drug combinations, and intervention timing. Earlier intervention in newly diagnosed HGG and avoidance of corticosteroids may improve efficacy in future trials. The results from ongoing trials demonstrate promising potential for molding the future landscape of HGG care.
Collapse
Affiliation(s)
- Matthew J Goldman
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Alexandra M Baskin
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA
- Houston Methodist Academic Institute, Houston, TX, USA
| | - Martyn A Sharpe
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA
- Houston Methodist Academic Institute, Houston, TX, USA
| | - David S Baskin
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA
- Houston Methodist Academic Institute, Houston, TX, USA
- Department of Neurosurgery, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Texas A & M Medical School
| |
Collapse
|
31
|
Zheng M, Wang Y, Chen S, Suo Y, Yu J, Zhang X. Enhancing Electric Field Distribution in the Pancreas for Improved TTFields Therapy: A Computational Modeling Investigation. IEEE Trans Biomed Eng 2024; 71:2612-2619. [PMID: 38564342 DOI: 10.1109/tbme.2024.3383818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
BACKGROUND Tumor treating fields (TTFields) therapy has shown effectiveness in glioblastoma treatment and holds potential for other cancers. However, its application in pancreatic cancer and the distribution of electric fields in pancreas remain unexplored. This study aims to investigate the electric field distributions in pancreatic regions using different array configurations for TTFields therapy. METHODS Computational modelling was employed to simulate electric field distributions, and quantitative analysis was conducted. Human body impedance measurements were used to optimize the electric properties of the model. Various array configurations were examined to assess their impact on the electric field distributions. RESULTS The study revealed that well-positioned arrays, specifically the combination of 20-piece transducer arrays in anterior-posterior orientation and 13-piece transducer arrays in left-right orientation, consistently achieved electric fields exceeding the 1V/cm threshold in over 99.4% of the pancreas. Even with a reduced number of transducers (13 pieces for both orientations), sufficient electric field coverage was achieved, exceeding the threshold in over 92.9% of the pancreas. Additionally, different array placements within the same orientation were explored to address clinical challenges such as skin rash and patient anatomical variations. CONCLUSIONS This research lays the groundwork for understanding TTFields distribution within the abdomen, offering insights into optimizing array configurations for improved electric field delivery. These results offer promises of advancing TTFields therapy for pancreatic cancer towards clinical applications, and potentially enhancing treatment efficacy and patient outcomes.
Collapse
|
32
|
INTERVAL-GB Collaborative, Gillespie CS, Bligh ER, Poon MTC, Islim AI, Solomou G, Gough M, Millward CP, Rominiyi O, Zakaria R, Price SJ, Watts C, Camp S, Booth TC, Thompson G, Mills SJ, Waldman A, Brennan PM, Jenkinson MD, Abdullmalek H, Abualsaud S, Adegboyega G, Afulukwe C, Ahmed N, Amoo M, Al-Sousi AN, Al-Tamimi Y, Anand A, Barua N, Bhatt H, Boiangiu I, Boyle A, Bredell C, Chaudri T, Cheong J, Cios A, Coope D, Coulter I, Critchley G, Davis H, De Luna PJ, Dey N, Duric B, Egiz A, Ekert JO, Egu CB, Ekanayake J, Elso A, Ferreira T, Flannery T, Fung KW, Ganguly R, Goyal S, Hardman E, Harris L, Hirst T, Hoah KS, Hodgson S, Hossain-Ibrahim K, Houlihan LM, Houssaini SS, Hoque S, Hutton D, Javed M, Kalra N, Kannan S, Kapasouri EM, Keenlyside A, Kehoe K, Kewlani B, Khanna P, de Koning R, Kumar KS, Kuri A, Lammy S, Lee E, Magouirk R, Martin AJ, Masina R, Mathew R, Mazzoleni A, McAleavey P, McKenna G, McSweeney D, Moughal S, Mustafa MA, Mthunzi E, Nazari A, Ngoc TTN, Nischal S, O’Sullivan M, Park JJ, Pandit AS, Smith JP, Peterson P, Phang I, Plaha P, Pujara S, Richardson GE, Saad M, et alINTERVAL-GB Collaborative, Gillespie CS, Bligh ER, Poon MTC, Islim AI, Solomou G, Gough M, Millward CP, Rominiyi O, Zakaria R, Price SJ, Watts C, Camp S, Booth TC, Thompson G, Mills SJ, Waldman A, Brennan PM, Jenkinson MD, Abdullmalek H, Abualsaud S, Adegboyega G, Afulukwe C, Ahmed N, Amoo M, Al-Sousi AN, Al-Tamimi Y, Anand A, Barua N, Bhatt H, Boiangiu I, Boyle A, Bredell C, Chaudri T, Cheong J, Cios A, Coope D, Coulter I, Critchley G, Davis H, De Luna PJ, Dey N, Duric B, Egiz A, Ekert JO, Egu CB, Ekanayake J, Elso A, Ferreira T, Flannery T, Fung KW, Ganguly R, Goyal S, Hardman E, Harris L, Hirst T, Hoah KS, Hodgson S, Hossain-Ibrahim K, Houlihan LM, Houssaini SS, Hoque S, Hutton D, Javed M, Kalra N, Kannan S, Kapasouri EM, Keenlyside A, Kehoe K, Kewlani B, Khanna P, de Koning R, Kumar KS, Kuri A, Lammy S, Lee E, Magouirk R, Martin AJ, Masina R, Mathew R, Mazzoleni A, McAleavey P, McKenna G, McSweeney D, Moughal S, Mustafa MA, Mthunzi E, Nazari A, Ngoc TTN, Nischal S, O’Sullivan M, Park JJ, Pandit AS, Smith JP, Peterson P, Phang I, Plaha P, Pujara S, Richardson GE, Saad M, Sangal S, Shanbhag A, Shetty V, Simon N, Spencer R, Sun R, Syed I, Sunny JT, Vasilica AM, O’Flaherty D, Raja A, Ramsay D, Reddi R, Roman E, Rominiyi O, Roy D, Salim O, Samkutty J, Selvakumar J, Santarius T, Smith S, Sofela A, St. George EJ, Subramanian P, Sundaresan V, Sweeney K, Tan BH, Turnbull N, Tao Y, Thorne L, Tweedie R, Tzatzidou A, Vaqas B, Venturini S, Whitehouse K, Whitfield P, Wildman J, Williams I, Williams K, Wykes V, Ye TTS, Yap KS, Yousuff M, Zulfiqar A, Neurology and Neurosurgery Interest Group (NANSIG), Bandyopadhyay S, Ooi SZY, Clynch A, Burton O, Steinruecke M, Bolton W, Touzet AY, Redpath H, Lee SH, Erhabor J, Mantle O, Gillespie CS, Bligh ES, British Neurosurgical Trainee Research Collaborative (BNTRC), Kolias A, Woodfield J, Chari A, Borchert R, Piper R, Fountain DM, Poon MTC, Islim AI. Imaging timing after surgery for glioblastoma: an evaluation of practice in Great Britain and Ireland (INTERVAL-GB)- a multi-centre, cohort study. J Neurooncol 2024; 169:517-529. [PMID: 39105956 PMCID: PMC11341661 DOI: 10.1007/s11060-024-04705-3] [Show More Authors] [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: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE Post-operative MRI is used to assess extent of resection, monitor treatment response and detect progression in high-grade glioma. However, compliance with accepted guidelines for follow-up MRI, and impact on management/outcomes is unclear. METHODS Multi-center, retrospective observational cohort study of patients with confirmed WHO grade 4 glioma (August 2018-February 2019) receiving oncological treatment. PRIMARY OBJECTIVE investigate follow-up MRI surveillance practice and compliance with recommendations from NICE (Post-operative scan < 72h, MRI every 3-6 months) and EANO (Post-operative scan < 48h, MRI every 3 months). RESULTS There were 754 patients from 26 neuro-oncology centers with a median age of 63 years (IQR 54-70), yielding 10,100 (median, 12.5/person, IQR 5.2-19.4) person-months of follow-up. Of patients receiving debulking surgery, most patients had post-operative MRI within 72 h of surgery (78.0%, N = 407/522), and within 48 h of surgery (64.2%, N = 335/522). The median number of subsequent follow-up MRI scans was 1 (IQR 0-4). Compliance with NICE and EANO recommendations for follow-up MRI was 52.8% (N = 398/754) and 24.9% (N = 188/754), respectively. On multivariable Cox regression analysis, increased time spent in recommended follow-up according to NICE guidelines was associated with longer OS (HR 0.56, 95% CI 0.46-0.66, P < 0.001), but not PFS (HR 0.93, 95% CI 0.79-1.10, P = 0.349). Increased time spent in recommended follow-up according to EANO guidelines was associated with longer OS (HR 0.54, 95% CI 0.45-0.63, P < 0.001) but not PFS (HR 0.99, 95% CI 0.84-1.16, P = 0.874). CONCLUSION Regular surveillance follow-up for glioblastoma is associated with longer OS. Prospective trials are needed to determine whether regular or symptom-directed MRI influences outcomes.
Collapse
|
33
|
Ius T, Somma T, Pasqualetti F, Berardinelli J, Vitulli F, Caccese M, Cella E, Cenciarelli C, Pozzoli G, Sconocchia G, Zeppieri M, Gerardo C, Caffo M, Lombardi G. Local therapy in glioma: An evolving paradigm from history to horizons (Review). Oncol Lett 2024; 28:440. [PMID: 39081966 PMCID: PMC11287108 DOI: 10.3892/ol.2024.14573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/14/2024] [Indexed: 08/02/2024] Open
Abstract
Despite the implementation of multimodal treatments after surgery, glioblastoma (GBM) remains an incurable disease, posing a significant challenge in neuro-oncology. In this clinical setting, local therapy (LT), a developing paradigm, has received significant interest over time due to its potential to overcome the drawbacks of conventional therapy options for GBM. The present review aimed to trace the historical development, highlight contemporary advances and provide insights into the future horizons of LT in GBM management. In compliance with the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols criteria, a systematic review of the literature on the role of LT in GBM management was conducted. A total of 2,467 potentially relevant articles were found and, after removal of duplicates, 2,007 studies were screened by title and abstract (Cohen's κ coefficient=0.92). Overall, it emerged that 15, 10 and 6 clinical studies explored the clinical efficiency of intraoperative local treatment modalities, local radiotherapy and local immunotherapy, respectively. GBM recurrences occur within 2 cm of the radiation field in 80% of cases, emphasizing the significant influence of local factors on recurrence. This highlights the urgent requirement for LT strategies. In total, three primary reasons have thus led to the development of numerous LT solutions in recent decades: i) Intratumoral implants allow the blood-brain barrier to be bypassed, resulting in limited systemic toxicity; ii) LT facilitates bridging therapy between surgery and standard treatments; and iii) given the complexity of GBM, targeting multiple components of the tumor microenvironment through ligands specific to various elements could have a synergistic effect in treatments. Considering the spatial and temporal heterogeneity of GBM, the disease prognosis could be significantly improved by a combination of therapeutic strategies in the era of precision medicine.
Collapse
Affiliation(s)
- Tamara Ius
- Unit of Neurosurgery, Head-Neck and Neurosciences Department, University Hospital of Udine, I-33100 Udine, Italy
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | | | - Jacopo Berardinelli
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | - Francesca Vitulli
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | - Mario Caccese
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
| | - Eugenia Cella
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
- Medical Oncology 2, San Martino Hospital-IRCCS, I-16131 Genoa Italy
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology, National Research Council, I-00133 Roma, Italy
| | - Giacomo Pozzoli
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, I-00168 Rome, Italy
| | - Giuseppe Sconocchia
- Institute of Translational Pharmacology, National Research Council, I-00133 Roma, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, I-33100 Udine, Italy
| | - Caruso Gerardo
- Unit of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University Hospital of Messina, I-98125 Messina, Italy
| | - Maria Caffo
- Unit of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University Hospital of Messina, I-98125 Messina, Italy
| | - Giuseppe Lombardi
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
| |
Collapse
|
34
|
Biedermann GB, Merrifield K, Lustgarten L. Case report: Safety of Tumor Treating Fields therapy with an implantable cardiac pacemaker in a patient with glioblastoma. Front Oncol 2024; 14:1441146. [PMID: 39239269 PMCID: PMC11374662 DOI: 10.3389/fonc.2024.1441146] [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: 06/06/2024] [Accepted: 07/23/2024] [Indexed: 09/07/2024] Open
Abstract
Tumor Treating Fields (TTFields) therapy is an anti-cancer treatment modality that is delivered noninvasively to the tumor site via skin-placed arrays. The therapy is US Food and Drug Administration (FDA) approved and Conformité Européenne (CE) marked for adults with newly diagnosed and recurrent glioblastoma (GBM) (grade 4 glioma in the European Union). To date, there are limited data on the safety and efficacy of TTFields therapy in patients with implanted cardiac pacemakers. Herein, we report a case of a 79-year-old male patient with GBM receiving TTFields therapy with a prior medical history of cardiac events necessitating a cardiac pacemaker. The patient presented to the emergency department in May 2021 with newly onset left-sided weakness along with seizures. Based on an initial evaluation and results of the initial computed tomography (CT) scans (May 2021), the patient was clinically diagnosed with a high-grade glioma which was later confirmed as IDH wildtype following a biopsy. He was treated with radiotherapy (40 Gy in 15 fractions), followed by adjuvant temozolomide (TMZ) (75 mg/m2). TTFields therapy was initiated alongside maintenance TMZ (150 mg/m2). Average TTFields therapy usage was 67% throughout the duration of treatment. Follow-up CT scans (February and May of 2022) indicated stable disease. CT scans in August 2022 showed an increase in size of a mass with heterogeneous contrast enhancement and the patient subsequently passed away in October 2022. The patient's last cardiac tests demonstrated that the pacemaker was operational with adequate cardiac function. This report suggests that TTFields therapy concomitant with an implanted electronic device may be safe in patients with GBM.
Collapse
Affiliation(s)
- Gregory B Biedermann
- Department of Radiation Oncology, University of Missouri, Columbia, MO, United States
| | - Kathleen Merrifield
- Department of Global Medical Safety, Novocure Inc., New York, NY, United States
| | | |
Collapse
|
35
|
Latzer P, Zelba H, Battke F, Reinhardt A, Shao B, Bartsch O, Rabsteyn A, Harter J, Schulze M, Okech T, Golf A, Kyzirakos-Feger C, Kayser S, Pieper N, Feldhahn M, Wünsche J, Seitz C, Hadaschik D, Garbe C, Hauser TK, la Fougère C, Biskup D, Brooke D, Parker D, Martens UM, Illerhaus G, Blumenthal DT, Merrell R, Lorenzo LS, Hidvégi M, de Robles P, Kebir S, Li WW, Li VW, Williams M, Miller AM, Kesari S, Castro M, Desjardins A, Ashley DM, Friedman HS, Wen PY, Neil EC, Iwamoto FM, Sipos B, Geletneky K, Zender L, Glas M, Reardon DA, Biskup S. A real-world observation of patients with glioblastoma treated with a personalized peptide vaccine. Nat Commun 2024; 15:6870. [PMID: 39127809 PMCID: PMC11316744 DOI: 10.1038/s41467-024-51315-8] [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: 02/29/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024] Open
Abstract
Current treatment outcome of patients with glioblastoma (GBM) remains poor. Following standard therapy, recurrence is universal with limited survival. Tumors from 173 GBM patients are analysed for somatic mutations to generate a personalized peptide vaccine targeting tumor-specific neoantigens. All patients were treated within the scope of an individual healing attempt. Among all vaccinated patients, including 70 treated prior to progression (primary) and 103 treated after progression (recurrent), the median overall survival from first diagnosis is 31.9 months (95% CI: 25.0-36.5). Adverse events are infrequent and are predominantly grade 1 or 2. A vaccine-induced immune response to at least one of the vaccinated peptides is detected in blood samples of 87 of 97 (90%) monitored patients. Vaccine-specific T-cell responses are durable in most patients. Significantly prolonged survival is observed for patients with multiple vaccine-induced T-cell responses (53 months) compared to those with no/low induced responses (27 months; P = 0.03). Altogether, our results highlight that the application of personalized neoantigen-targeting peptide vaccine is feasible and represents a promising potential treatment option for GBM patients.
Collapse
Affiliation(s)
| | - Henning Zelba
- Zentrum für Humangenetik Tübingen, Tübingen, Germany
| | | | | | - Borong Shao
- Zentrum für Humangenetik Tübingen, Tübingen, Germany
| | | | | | | | | | - Thomas Okech
- MVZ Zentrum für ambulante Onkologie GmbH, Tübingen, Germany
| | - Alexander Golf
- MVZ Zentrum für ambulante Onkologie GmbH, Tübingen, Germany
| | | | - Simone Kayser
- Zentrum für Humangenetik Tübingen, Tübingen, Germany
| | | | | | - Julian Wünsche
- Zentrum für Humangenetik Tübingen, Tübingen, Germany
- MVZ Zentrum für ambulante Onkologie GmbH, Tübingen, Germany
| | - Christian Seitz
- Universitätsklinikum Heidelberg, KiTZ, Hopp Children's Cancer Center, Heidelberg, Germany
| | | | - Claus Garbe
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | | | - Christian la Fougère
- Nuklearmedizin und Klinische Molekulare Bildgebung, Universitätsklinikum Tübingen, Tübingen, Germany
| | | | - Dawn Brooke
- Provenance Precision Medicine Foundation, Wilmette, IL, USA
| | | | - Uwe M Martens
- Department of Hematology and Oncology, Cancer Center Heilbronn-Franken, SLK Kliniken GmbH, Heilbronn, Germany
| | | | | | - Ryan Merrell
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Máté Hidvégi
- Jewish Theological Seminary-University of Jewish Studies (OR-ZSE), Budapest, Hungary
| | - Paula de Robles
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Sied Kebir
- Universitätsklinikum Essen, Essen, Germany
| | | | - Vincent W Li
- Angiogenesis Foundation, Cambridge, MA, USA
- Harvard Medical School Dermatology, Boston, MA, USA
| | | | - Alexandra M Miller
- Brain and Spine Tumor Center, NYU Langone Health's Perlmutter Cancer Center, New York, NY, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Michael Castro
- Cellworks Group Inc, South San Francisco, CA, USA
- Personalized Cancer Medicine, PLLC, Santa Monica, CA, USA
| | - Annick Desjardins
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - David M Ashley
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Preston Robert Tisch Brain Tumor Centre, Duke University, Durham, NC, USA
| | - Henry S Friedman
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Elisabeth C Neil
- Department of Neurology, University of Minnesota Health, Minneapolis, MN, USA
| | | | - Bence Sipos
- BAG für Pathologie und Molekularpathologie Stuttgart, Molekularpathologie Baden-Württemberg GbR, Stuttgart, Germany
| | | | - Lars Zender
- University Hospital Tübingen, Internal Medicine VIII, Tübingen, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology, Universitätsklinikum Essen Klinik für Neurologie, Essen, Germany
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saskia Biskup
- Zentrum für Humangenetik Tübingen, Tübingen, Germany.
- CeGaT GmbH, Tübingen, Germany.
| |
Collapse
|
36
|
Dillman RO, Bota DA. Next-generation vaccines are showing promise against glioblastoma. Oncotarget 2024; 15:543-548. [PMID: 39102214 PMCID: PMC11299660 DOI: 10.18632/oncotarget.28636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Indexed: 08/06/2024] Open
Affiliation(s)
| | - Daniela A. Bota
- Correspondence to:Daniela A. Bota, Chao Family Comprehensive Cancer Center, Departments of Neurology and Neurological Surgery, University of California, Irvine, Orange, CA 92868, USA email
| |
Collapse
|
37
|
Akimoto T, Islam MR, Nagasako A, Kishi K, Nakakaji R, Ohtake M, Hasumi H, Yamaguchi T, Yamada S, Yamamoto T, Ishikawa Y, Umemura M. Alternative magnetic field exposure suppresses tumor growth via metabolic reprogramming. Cancer Sci 2024; 115:2686-2700. [PMID: 38877783 PMCID: PMC11309929 DOI: 10.1111/cas.16243] [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: 02/07/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
Application of physical forces, ranging from ultrasound to electric fields, is recommended in various clinical practice guidelines, including those for treating cancers and bone fractures. However, the mechanistic details of such treatments are often inadequately understood, primarily due to the absence of comprehensive study models. In this study, we demonstrate that an alternating magnetic field (AMF) inherently possesses a direct anti-cancer effect by enhancing oxidative phosphorylation (OXPHOS) and thereby inducing metabolic reprogramming. We observed that the proliferation of human glioblastoma multiforme (GBM) cells (U87 and LN229) was inhibited upon exposure to AMF within a specific narrow frequency range, including around 227 kHz. In contrast, this exposure did not affect normal human astrocytes (NHA). Additionally, in mouse models implanted with human GBM cells in the brain, daily exposure to AMF for 30 min over 21 days significantly suppressed tumor growth and prolonged overall survival. This effect was associated with heightened reactive oxygen species (ROS) production and increased manganese superoxide dismutase (MnSOD) expression. The anti-cancer efficacy of AMF was diminished by either a mitochondrial complex IV inhibitor or a ROS scavenger. Along with these observations, there was a decrease in the extracellular acidification rate (ECAR) and an increase in the oxygen consumption rate (OCR). This suggests that AMF-induced metabolic reprogramming occurs in GBM cells but not in normal cells. Our results suggest that AMF exposure may offer a straightforward strategy to inhibit cancer cell growth by leveraging oxidative stress through metabolic reprogramming.
Collapse
Affiliation(s)
- Taisuke Akimoto
- Department of NeurosurgeryYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Md Rafikul Islam
- Department of Biochemistry and Molecular BiologyWinthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences (UAMS)Little RockArkansasUSA
| | - Akane Nagasako
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | | | - Rina Nakakaji
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Makoto Ohtake
- Department of NeurosurgeryYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Hisashi Hasumi
- Department of Urology, Yokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | | | - Shigeki Yamada
- Department of Materials System Science, Yokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Tetsuya Yamamoto
- Department of NeurosurgeryYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Yoshihiro Ishikawa
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| | - Masanari Umemura
- Cardiovascular Research InstituteYokohama City University Graduate School of MedicineYokohamaKanagawaJapan
| |
Collapse
|
38
|
Mrugala MM, Shi W, Iwomoto F, Lukas RV, Palmer JD, Suh JH, Glas M. Global post‑marketing safety surveillance of Tumor Treating Fields (TTFields) therapy in over 25,000 patients with CNS malignancies treated between 2011-2022. J Neurooncol 2024; 169:25-38. [PMID: 38949692 PMCID: PMC11269345 DOI: 10.1007/s11060-024-04682-7] [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: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Tumor Treating Fields (TTFields) are alternating electric fields that disrupt cancer cell processes. TTFields therapy is approved for recurrent glioblastoma (rGBM), and newly-diagnosed (nd) GBM (with concomitant temozolomide for ndGBM; US), and for grade IV glioma (EU). We present an updated global, post-marketing surveillance safety analysis of patients with CNS malignancies treated with TTFields therapy. METHODS Safety data were collected from routine post-marketing activities for patients in North America, Europe, Israel, and Japan (October 2011-October 2022). Adverse events (AEs) were stratified by age, sex, and diagnosis. RESULTS Overall, 25,898 patients were included (diagnoses: ndGBM [68%], rGBM [26%], anaplastic astrocytoma/oligodendroglioma [4%], other CNS malignancies [2%]). Median (range) age was 59 (3-103) years; 66% patients were male. Most (69%) patients were 18-65 years; 0.4% were < 18 years; 30% were > 65 years. All-cause and TTFields-related AEs occurred in 18,798 (73%) and 14,599 (56%) patients, respectively. Most common treatment-related AEs were beneath-array skin reactions (43%), electric sensation (tingling; 14%), and heat sensation (warmth; 12%). Treatment-related skin reactions were comparable in pediatric (39%), adult (42%), and elderly (45%) groups, and in males (41%) and females (46%); and similar across diagnostic subgroups (ndGBM, 46%; rGBM, 34%; anaplastic astrocytoma/oligodendroglioma, 42%; other, 40%). No TTFields-related systemic AEs were reported. CONCLUSIONS This long-term, real-world analysis of > 25,000 patients demonstrated good tolerability of TTFields in patients with CNS malignancies. Most therapy-related AEs were manageable localized, non-serious skin events. The TTFields therapy safety profile remained consistent across subgroups (age, sex, and diagnosis), indicative of its broad applicability.
Collapse
Affiliation(s)
- Maciej M Mrugala
- Mayo Clinic College of Medicine and Science, Mayo Clinic, Phoenix/Scottsdale, Arizona, USA.
| | - Wenyin Shi
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Fabio Iwomoto
- Division of Neuro-Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY, USA
| | - Rimas V Lukas
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Joshua D Palmer
- The Department of Radiation Oncology, The James Cancer Hospital, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - John H Suh
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, West German Cancer Center (WTZ) and German Cancer Consortium, Partner Site, Essen, Germany
| |
Collapse
|
39
|
Zhao X, Jakobsson V, Tao Y, Zhao T, Wang J, Khong PL, Chen X, Zhang J. Targeted Radionuclide Therapy in Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39042829 DOI: 10.1021/acsami.4c07850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Despite the development of various novel therapies, glioblastoma (GBM) remains a devastating disease, with a median survival of less than 15 months. Recently, targeted radionuclide therapy has shown significant progress in treating solid tumors, with the approval of Lutathera for neuroendocrine tumors and Pluvicto for prostate cancer by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This achievement has shed light on the potential of targeted radionuclide therapy for other solid tumors, including GBM. This review presents the current status of targeted radionuclide therapy in GBM, highlighting the commonly used therapeutic radionuclides emitting alpha, beta particles, and Auger electrons that could induce potent molecular and cellular damage to treat GBM. We then explore a range of targeting vectors, including small molecules, peptides, and antibodies, which selectively target antigen-expressing tumor cells with minimal or no binding to healthy tissues. Considering that radiopharmaceuticals for GBM are often administered locoregionally to bypass the blood-brain barrier (BBB), we review prominent delivery methods such as convection-enhanced delivery, local implantation, and stereotactic injections. Finally, we address the challenges of this therapeutic approach for GBM and propose potential solutions.
Collapse
Affiliation(s)
- Xiaobin Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yucen Tao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingyan Wang
- Xiamen University, School of Public Health, Xiang'an South Road, Xiamen 361102, China
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
40
|
Han W, Zhou H, Zhang X, Li H, Han X, Su L, Tian L, Xue X. HMGB2 is a biomarker associated with poor prognosis promoting radioresistance in glioma by targeting base excision repair pathway. Transl Oncol 2024; 45:101977. [PMID: 38728871 PMCID: PMC11107350 DOI: 10.1016/j.tranon.2024.101977] [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: 11/29/2023] [Revised: 03/27/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND High mobility group box 2 (HMGB2) is considered as a biomarker of poor prognosis in various cancers.This study aims to investigate the effect and mechanism of HMGB2 in gliomas. METHODS With the glioma related on-line and our local hospital databases, the expression differences of HMGB2,Kaplan-Meier survival analysis and COX regression analysis were performed.The correlation analysis between the clinicopathological features and imaging parameters with the HMGB2 expression had been done. Then GSEA and PPI networks were carried out to find out the most significant pathway. The pathway inhibitor was applied to verify HMGB2's participation. CCK8,EDU assays,γ-H2AX immunofluorescence staining and colony formation assay were conducted to observe effects on glioma cells. RESULTS Available datasets showed that HMGB2 was highly expressed in glioma and patients with high expression of HMGB2 had poorer prognosis and molecular characteristics. Protein level evidence of western blot and immunohistochemistry from our center supported the conclusions above. Analysis on imaging features suggested that HMGB2 expression level had an inverse association with ADCmean but positively with the thickness of enhancing margin. Results from GSEA and PPI network analysis exhibited that HMGB2 was involved in base excision repair (BER) signaling pathway. Experimental evidence demonstrated that the overexpression of HMGB2 promoted the proliferation of glioma cells and enhanced the radio-resistance. CONCLUSIONS HMGB2 could promote glioma development and enhance the radioresistance of glioma cells, potentially related to the BER pathway, suggesting it may serve as an underlying biomarker for patients with glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Huandi Zhou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinyuan Zhang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Haonan Li
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuetao Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Linlin Su
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Tian
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoying Xue
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
41
|
Sabri ME, Moghaddasi L, Wilson P, Saran F, Bezak E. Targeted Alpha Therapy for Glioblastoma: Review on In Vitro, In Vivo and Clinical Trials. Target Oncol 2024; 19:511-531. [PMID: 38836953 PMCID: PMC11230998 DOI: 10.1007/s11523-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
Glioblastoma (GB), a prevalent and highly malignant primary brain tumour with a very high mortality rate due to its resistance to conventional therapies and invasive nature, resulting in 5-year survival rates of only 4-17%. Despite recent advancements in cancer management, the survival rates for GB patients have not significantly improved over the last 10-20 years. Consequently, there exists a critical unmet need for innovative therapies. One promising approach for GB is Targeted Alpha Therapy (TAT), which aims to selectively deliver potentially therapeutic radiation doses to malignant cells and the tumour microenvironment while minimising radiation exposure to surrounding normal tissue with or without conventional external beam radiation. This approach has shown promise in both pre-clinical and clinical settings. A review was conducted following PRISMA 2020 guidelines across Medline, SCOPUS, and Embase, identifying 34 relevant studies out of 526 initially found. In pre-clinical studies, TAT demonstrated high binding specificity to targeted GB cells, with affinity rates between 60.0% and 84.2%, and minimal binding to non-targeted cells (4.0-5.6%). This specificity significantly enhanced cytotoxic effects and improved biodistribution when delivered intratumorally. Mice treated with TAT showed markedly higher median survival rates compared to control groups. In clinical trials, TAT applied to recurrent GB (rGB) displayed varying success rates in extending overall survival (OS) and progression-free survival. Particularly effective when integrated into treatment regimens for both newly diagnosed and recurrent cases, TAT increased the median OS by 16.1% in newly diagnosed GB and by 36.4% in rGB, compared to current standard therapies. Furthermore, it was generally well tolerated with minimal adverse effects. These findings underscore the potential of TAT as a viable therapeutic option in the management of GB.
Collapse
Affiliation(s)
- Maram El Sabri
- Allied Health and Human Performance, University of South Australia, University of South Australia City East Campus, Adelaide, SA, 5001, Australia.
| | - Leyla Moghaddasi
- Department of Medical Physics, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Puthenparampil Wilson
- UniSA STEM, University of South Australia, Adelaide, SA, 5001, Australia
- Department of Medical Physics, Royal Adelaide Hospital, Adelaide, Australia
| | - Frank Saran
- Allied Health and Human Performance, University of South Australia, University of South Australia City East Campus, Adelaide, SA, 5001, Australia
- Australian Bragg Centre for Proton Therapy and Research, Adelaide, SA, 5000, Australia
- Department of Radiotherapy, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Eva Bezak
- Allied Health and Human Performance, University of South Australia, University of South Australia City East Campus, Adelaide, SA, 5001, Australia
| |
Collapse
|
42
|
Cao X, Li J, Ren J, Peng J, Zhong R, He J, Xu T, Yu Z, Jin H, Hao S, Liu R, Xu B. Minimally-invasive implantable device enhances brain cancer suppression. EMBO Mol Med 2024; 16:1704-1716. [PMID: 38902433 PMCID: PMC11250787 DOI: 10.1038/s44321-024-00091-5] [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: 02/20/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
Current brain tumor treatments are limited by the skull and BBB, leading to poor prognosis and short survival for glioma patients. We introduce a novel minimally-invasive brain tumor suppression (MIBTS) device combining personalized intracranial electric field therapy with in-situ chemotherapeutic coating. The core of our MIBTS technique is a wireless-ultrasound-powered, chip-sized, lightweight device with all functional circuits encapsulated in a small but efficient "Swiss-roll" structure, guaranteeing enhanced energy conversion while requiring tiny implantation windows ( ~ 3 × 5 mm), which favors broad consumers acceptance and easy-to-use of the device. Compared with existing technologies, competitive advantages in terms of tumor suppressive efficacy and therapeutic resolution were noticed, with maximum ~80% higher suppression effect than first-line chemotherapy and 50-70% higher than the most advanced tumor treating field technology. In addition, patient-personalized therapy strategies could be tuned from the MIBTS without increasing size or adding circuits on the integrated chip, ensuring the optimal therapeutic effect and avoid tumor resistance. These groundbreaking achievements of MIBTS offer new hope for controlling tumor recurrence and extending patient survival.
Collapse
Affiliation(s)
- Xiaona Cao
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jie Li
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jinliang Ren
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jiajin Peng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Ruyue Zhong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Jiahao He
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Ting Xu
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China
| | - Zhenhua Yu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Huawei Jin
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Siqi Hao
- School of Naval Architecture & Ocean Engineering, Guangzhou Maritime University, 101 Hongshan 3rd Road, Huangpu District, Guangzhou, Guangdong, 510725, P.R. China
| | - Ruiwei Liu
- School of Naval Architecture & Ocean Engineering, Guangzhou Maritime University, 101 Hongshan 3rd Road, Huangpu District, Guangzhou, Guangdong, 510725, P.R. China
| | - Bingzhe Xu
- School of Biomedical Engineering, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou, 510275, P.R. China.
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, 518107, P.R. China.
| |
Collapse
|
43
|
Berckmans Y, Ene HM, Ben-Meir K, Martinez-Conde A, Wouters R, Van den Ende B, Van Mechelen S, Monin R, Frechtel-Gerzi R, Gabay H, Dor-On E, Haber A, Weinberg U, Vergote I, Giladi M, Coosemans A, Palti Y. Tumor Treating Fields (TTFields) induce homologous recombination deficiency in ovarian cancer cells, thus mitigating drug resistance. Front Oncol 2024; 14:1402851. [PMID: 38993641 PMCID: PMC11238040 DOI: 10.3389/fonc.2024.1402851] [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: 03/18/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
Background Ovarian cancer is the leading cause of mortality among gynecological malignancies. Carboplatin and poly (ADP-ribose) polymerase inhibitors (PARPi) are often implemented in the treatment of ovarian cancer. Homologous recombination deficient (HRD) tumors demonstrate increased sensitivity to these treatments; however, many ovarian cancer patients are homologous recombination proficient (HRP). TTFields are non-invasive electric fields that induce an HRD-like phenotype in various cancer types. The current study aimed to investigate the impact of TTFields applied together with carboplatin or PARPi (olaparib or niraparib) in preclinical ovarian cancer models. Methods A2780 (HRP), OVCAR3 (HRD), and A2780cis (platinum-resistant) human ovarian cancer cells were treated in vitro with TTFields (1 V/cm RMS, 200 kHz, 72 h), alone or with various drug concentrations. Treated cells were measured for cell count, colony formation, apoptosis, DNA damage, expression of DNA repair proteins, and cell cycle. In vivo, ID8-fLuc (HRP) ovarian cancer cells were inoculated intraperitoneally to C57BL/6 mice, which were then treated with either sham, TTFields (200 kHz), olaparib (50 mg/kg), or TTFields plus olaparib; over a period of four weeks. Tumor growth was analyzed using bioluminescent imaging at treatment cessation; and survival analysis was performed. Results The nature of TTFields-drug interaction was dependent on the drug's underlying mechanism of action and on the genetic background of the cells, with synergistic interactions between TTFields and carboplatin or PARPi seen in HRP and resistant cells. Treated cells demonstrated elevated levels of DNA damage, accompanied by G2/M arrest, and induction of an HRD-like phenotype. In the tumor-bearing mice, TTFields and olaparib co-treatment resulted in reduced tumor volume and a survival benefit relative to olaparib monotherapy and to control. Conclusion By inducing an HRD-like phenotype, TTFields sensitize HRP and resistant ovarian cancer cells to treatment with carboplatin or PARPi, potentially mitigating a-priori and de novo drug resistance, a major limitation in ovarian cancer treatment.
Collapse
Affiliation(s)
- Yani Berckmans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | | | | | - Roxanne Wouters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
- Oncoinvent AS, Oslo, Norway
| | - Bieke Van den Ende
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sara Van Mechelen
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | - Ignace Vergote
- Department of Gynecology and Obstetrics, Gynecologic Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
44
|
Chang C, Chavarro VS, Gerstl JVE, Blitz SE, Spanehl L, Dubinski D, Valdes PA, Tran LN, Gupta S, Esposito L, Mazzetti D, Gessler FA, Arnaout O, Smith TR, Friedman GK, Peruzzi P, Bernstock JD. Recurrent Glioblastoma-Molecular Underpinnings and Evolving Treatment Paradigms. Int J Mol Sci 2024; 25:6733. [PMID: 38928445 PMCID: PMC11203521 DOI: 10.3390/ijms25126733] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma is the most common and lethal central nervous system malignancy with a median survival after progression of only 6-9 months. Major biochemical mechanisms implicated in glioblastoma recurrence include aberrant molecular pathways, a recurrence-inducing tumor microenvironment, and epigenetic modifications. Contemporary standard-of-care (surgery, radiation, chemotherapy, and tumor treating fields) helps to control the primary tumor but rarely prevents relapse. Cytoreductive treatment such as surgery has shown benefits in recurrent glioblastoma; however, its use remains controversial. Several innovative treatments are emerging for recurrent glioblastoma, including checkpoint inhibitors, chimeric antigen receptor T cell therapy, oncolytic virotherapy, nanoparticle delivery, laser interstitial thermal therapy, and photodynamic therapy. This review seeks to provide readers with an overview of (1) recent discoveries in the molecular basis of recurrence; (2) the role of surgery in treating recurrence; and (3) novel treatment paradigms emerging for recurrent glioblastoma.
Collapse
Affiliation(s)
- Christopher Chang
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
| | - Velina S. Chavarro
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
| | - Jakob V. E. Gerstl
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
| | - Sarah E. Blitz
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Lennard Spanehl
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Department of Neurosurgery, University of Rostock, 18055 Rostock, Germany; (D.D.); (F.A.G.)
| | - Daniel Dubinski
- Department of Neurosurgery, University of Rostock, 18055 Rostock, Germany; (D.D.); (F.A.G.)
| | - Pablo A. Valdes
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Lily N. Tran
- Division of Biology and Medicine, Brown University, Providence, RI 02912, USA;
| | - Saksham Gupta
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Luisa Esposito
- Department of Medicine and Surgery, Unicamillus University, 00131 Rome, Italy;
| | - Debora Mazzetti
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
| | - Florian A. Gessler
- Department of Neurosurgery, University of Rostock, 18055 Rostock, Germany; (D.D.); (F.A.G.)
| | - Omar Arnaout
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Timothy R. Smith
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Gregory K. Friedman
- Division of Pediatrics, Neuro-Oncology Section, MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Pierpaolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA; (V.S.C.); (J.V.E.G.); (S.E.B.); (L.S.); (S.G.); (D.M.); (O.A.); (T.R.S.); (J.D.B.)
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
45
|
White J, White MPJ, Wickremesekera A, Peng L, Gray C. The tumour microenvironment, treatment resistance and recurrence in glioblastoma. J Transl Med 2024; 22:540. [PMID: 38844944 PMCID: PMC11155041 DOI: 10.1186/s12967-024-05301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024] Open
Abstract
The adaptability of glioblastoma (GBM) cells, encouraged by complex interactions with the tumour microenvironment (TME), currently renders GBM an incurable cancer. Despite intensive research, with many clinical trials, GBM patients rely on standard treatments including surgery followed by radiation and chemotherapy, which have been observed to induce a more aggressive phenotype in recurrent tumours. This failure to improve treatments is undoubtedly a result of insufficient models which fail to incorporate components of the human brain TME. Research has increasingly uncovered mechanisms of tumour-TME interactions that correlate to worsened patient prognoses, including tumour-associated astrocyte mitochondrial transfer, neuronal circuit remodelling and immunosuppression. This tumour hijacked TME is highly implicated in driving therapy resistance, with further alterations within the TME and tumour resulting from therapy exposure inducing increased tumour growth and invasion. Recent developments improving organoid models, including aspects of the TME, are paving an exciting future for the research and drug development for GBM, with the hopes of improving patient survival growing closer. This review focuses on GBMs interactions with the TME and their effect on tumour pathology and treatment efficiency, with a look at challenges GBM models face in sufficiently recapitulating this complex and highly adaptive cancer.
Collapse
Affiliation(s)
- Jasmine White
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | | | - Agadha Wickremesekera
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
46
|
Zhang L, Ren Y, Peng Y, Luo Y, Liu Y, Wang X, Yang Y, Liu L, Ai P, Yang X, Li Y, Mao Q, Wang F. Tumor treating fields for newly diagnosed high-grade glioma based on the criteria of 2021 WHO CNS5: A retrospective analysis of Chinese patients in a single center. Cancer Med 2024; 13:e7350. [PMID: 38859683 PMCID: PMC11165168 DOI: 10.1002/cam4.7350] [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: 11/10/2023] [Revised: 04/04/2024] [Accepted: 05/26/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVE High-grade glioma (HGG) is known to be characterized by a high degree of malignancy and a worse prognosis. The classical treatment is safe resection supplemented by radiotherapy and chemotherapy. Tumor treating fields (TTFields), an emerging physiotherapeutic modality that targets malignant solid tumors using medium-frequency, low-intensity, alternating electric fields to interfere with cell division, have been used for the treatment of new diagnosis of glioblastoma, however, their administration in HGG requires further clinical evidence. The efficacy and safety of TTFields in Chinese patients with HGG were retrospectively evaluated by us in a single center. METHODS We enrolled and analyzed 52 patients with newly diagnosed HGG undergoing surgery and standard chemoradiotherapy regimens from December 2019 to June 2022, and followed them until June 2023. Based on whether they used TTFields, they were divided into a TTFields group and a non-TTFields group. Progression-free survival (PFS) and overall survival (OS) were compared between the two groups. RESULTS There were 26 cases in the TTFields group and 26 cases in the non-TTFields group. In the TTFields group, the median PFS was 14.2 months (95% CI: 9.50-18.90), the median OS was 19.7 months (95% CI: 14.95-24.25) , the median interval from surgery to the start of treatment with TTFields was 2.47 months (95% CI: 1.47-4.13), and the median duration of treatment with TTFields was 10.6 months (95% CI: 9.57-11.63). 15 (57.69%) patients experienced an adverse event and no serious adverse event was reported. In the non-TTFields group, the median PFS was 9.57 months (95% CI: 6.23-12.91) and the median OS was 16.07 months (95% CI: 12.90-19.24). There was a statistically significant difference in PFS (p = 0.005) and OS (p = 0.007) between the two groups. CONCLUSIONS In this retrospective analysis, TTFields were observed to improve newly diagnosed HGG patients' median PFS and OS. Compliance was much higher than reported in clinical trials and safety remained good.
Collapse
Affiliation(s)
- Li Zhang
- Head and Neck Oncology Ward, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Yanming Ren
- Department of Neurosurgery, West China HospitalSichuan UniversityChengduChina
| | - Youheng Peng
- Department of Neurosurgery, West China HospitalSichuan UniversityChengduChina
| | - Yong Luo
- Head and Neck Oncology Ward, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Yanhui Liu
- Department of Neurosurgery, West China HospitalSichuan UniversityChengduChina
| | - Xiang Wang
- Department of Neurosurgery, West China HospitalSichuan UniversityChengduChina
| | - Yuan Yang
- Department of Neurosurgery, West China HospitalSichuan UniversityChengduChina
| | - Lei Liu
- Head and Neck Oncology Ward, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Ping Ai
- Head and Neck Oncology Ward, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Xiaoyan Yang
- Head and Neck Oncology Ward, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Yanchu Li
- Head and Neck Oncology Ward, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Qing Mao
- Department of Neurosurgery, West China HospitalSichuan UniversityChengduChina
| | - Feng Wang
- Head and Neck Oncology Ward, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
47
|
Chen J, Liu Y, Lan J, Liu H, Tang Q, Li Z, Qiu X, Hu W, Xie J, Feng Y, Qin L, Zhang X, Liu J, Chen L. Identification and validation of COL6A1 as a novel target for tumor electric field therapy in glioblastoma. CNS Neurosci Ther 2024; 30:e14802. [PMID: 38887185 PMCID: PMC11183175 DOI: 10.1111/cns.14802] [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: 01/25/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most aggressive primary brain malignancy. Novel therapeutic modalities like tumor electric field therapy (TEFT) have shown promise, but underlying mechanisms remain unclear. The extracellular matrix (ECM) is implicated in GBM progression, warranting investigation into TEFT-ECM interplay. METHODS T98G cells were treated with TEFT (200 kHz, 2.2 V/m) for 72 h. Collagen type VI alpha 1 (COL6A1) was identified as hub gene via comprehensive bioinformatic analysis based on RNA sequencing (RNA-seq) and public glioma datasets. TEFT intervention models were established using T98G and Ln229 cell lines. Pre-TEFT and post-TEFT GBM tissues were collected for further validation. Focal adhesion pathway activity was assessed by western blot. Functional partners of COL6A1 were identified and validated by co-localization and survival analysis. RESULTS TEFT altered ECM-related gene expression in T98G cells, including the hub gene COL6A1. COL6A1 was upregulated in GBM and associated with poor prognosis. Muti-database GBM single-cell analysis revealed high-COL6A1 expression predominantly in malignant cell subpopulations. Differential expression and functional enrichment analyses suggested COL6A1 might be involved in ECM organization and focal adhesion. Western blot (WB), immunofluorescence (IF), and co-immunoprecipitation (Co-IP) experiments revealed that TEFT significantly inhibited expression of COL6A1, hindering its interaction with ITGA5, consequently suppressing the FAK/Paxillin/AKT pathway activity. These results suggested that TEFT might exert its antitumor effects by downregulating COL6A1 and thereby inhibiting the activity of the focal adhesion pathway. CONCLUSION TEFT could remodel the ECM of GBM cells by downregulating COL6A1 expression and inhibiting focal adhesion pathway. COL6A1 could interact with ITGA5 and activate the focal adhesion pathway, suggesting that it might be a potential therapeutic target mediating the antitumor effects of TEFT.
Collapse
Affiliation(s)
- Junyi Chen
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| | - Yuyang Liu
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jinxin Lan
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
| | - Hongyu Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryHainan Hospital of Chinese PLA General HospitalHainanChina
| | - Qingyun Tang
- Department of Gastroenterology920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Ze Li
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| | - Xiaoguang Qiu
- Beijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Wentao Hu
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
| | - Jiaxin Xie
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Yaping Feng
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Lilin Qin
- Zhejiang Cancer HospitalZhejiangHangzhouChina
| | - Xin Zhang
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jialin Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| | - Ling Chen
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
| |
Collapse
|
48
|
Qin X, Shi H, Wen Z, Chu B, Li H, Wang H, He Y, Sun X. Triboelectric-Responsive Drug Delivery Hydrogel for Accelerating Infected Wound Healing. Adv Healthc Mater 2024; 13:e2303474. [PMID: 38458151 DOI: 10.1002/adhm.202303474] [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: 10/11/2023] [Revised: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Electrotherapy is of great interest in the field of tissue repair as an effective, well-tolerated, and noninvasive treatment. Triboelectric nanogenerator (TENG) has shown advantages in promoting wound healing due to its peak output characteristic and low Joule heating effect. However, it is limited in infected wound healing due to poor antimicrobial capacity. Here, a wearable triboelectric stimulator (WTS) is developed that consists of a flexible TENG (F-TENG) and a triboelectric-responsive drug delivery hydrogel (TR-DDH) for healing of bacterium-infected wounds. F-TENG can generate pulsed current to wounds by converting mechanical energy from body movements. Polypyrrole is prone to reduction and volume contraction under electrical stimulation, resulting in desorption of curcumin nanoparticles (CUR NPs) from the polypyrrole in TR-DDH. Therefore, the highly efficient and controllable release of CUR NPs can be achieved by triboelectric stimulation. According to the in vitro and in vivo experiments, WTS has the greatest antimicrobial effect and the fastest promotion of infected wound healing compared to treatment with electrical stimulation or curcumin. Finally, the safety assessment demonstrates that the WTS has excellent tissue safety for chronic wound healing. Synergistic therapy with WTS provides an efficient strategy for chronic wound healing and smart-responsive drug delivery systems.
Collapse
Affiliation(s)
- Xuan Qin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Haoliang Shi
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Zhen Wen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Binbin Chu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Hongyang Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Houyu Wang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Yao He
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering (MIMSE), Macau University of Science and Technology, Macau, 999078, China
| | - Xuhui Sun
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
49
|
Sadowski K, Jażdżewska A, Kozłowski J, Zacny A, Lorenc T, Olejarz W. Revolutionizing Glioblastoma Treatment: A Comprehensive Overview of Modern Therapeutic Approaches. Int J Mol Sci 2024; 25:5774. [PMID: 38891962 PMCID: PMC11172387 DOI: 10.3390/ijms25115774] [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: 05/05/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in the adult population, with an average survival of 12.1 to 14.6 months. The standard treatment, combining surgery, radiotherapy, and chemotherapy, is not as efficient as we would like. However, the current possibilities are no longer limited to the standard therapies due to rapid advancements in biotechnology. New methods enable a more precise approach by targeting individual cells and antigens to overcome cancer. For the treatment of glioblastoma, these are gamma knife therapy, proton beam therapy, tumor-treating fields, EGFR and VEGF inhibitors, multiple RTKs inhibitors, and PI3K pathway inhibitors. In addition, the increasing understanding of the role of the immune system in tumorigenesis and the ability to identify tumor-specific antigens helped to develop immunotherapies targeting GBM and immune cells, including CAR-T, CAR-NK cells, dendritic cells, and immune checkpoint inhibitors. Each of the described methods has its advantages and disadvantages and faces problems, such as the inefficient crossing of the blood-brain barrier, various neurological and systemic side effects, and the escape mechanism of the tumor. This work aims to present the current modern treatments of glioblastoma.
Collapse
Affiliation(s)
- Karol Sadowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Adrianna Jażdżewska
- The Department of Anatomy and Neurobiology, Medical University of Gdansk, Dębinki 1, 80-211 Gdansk, Poland;
| | - Jan Kozłowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Aleksandra Zacny
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Tomasz Lorenc
- Department of Radiology I, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
50
|
Smerdi D, Moutafi M, Kotsantis I, Stavrinou LC, Psyrri A. Overcoming Resistance to Temozolomide in Glioblastoma: A Scoping Review of Preclinical and Clinical Data. Life (Basel) 2024; 14:673. [PMID: 38929657 PMCID: PMC11204771 DOI: 10.3390/life14060673] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma (GB) is the most common and most aggressive primary brain tumor in adults, with an overall survival almost 14.6 months. Optimal resection followed by combined temozolomide chemotherapy and radiotherapy, also known as Stupp protocol, remains the standard of treatment; nevertheless, resistance to temozolomide, which can be obtained throughout many molecular pathways, is still an unsurpassed obstacle. Several factors influence the efficacy of temozolomide, including the involvement of other DNA repair systems, aberrant signaling pathways, autophagy, epigenetic modifications, microRNAs, and extracellular vesicle production. The blood-brain barrier, which serves as both a physical and biochemical obstacle, the tumor microenvironment's pro-cancerogenic and immunosuppressive nature, and tumor-specific characteristics such as volume and antigen expression, are the subject of ongoing investigation. In this review, preclinical and clinical data about temozolomide resistance acquisition and possible ways to overcome chemoresistance, or to treat gliomas without restoration of chemosensitinity, are evaluated and presented. The objective is to offer a thorough examination of the clinically significant molecular mechanisms and their intricate interrelationships, with the aim of enhancing understanding to combat resistance to TMZ more effectively.
Collapse
Affiliation(s)
- Dimitra Smerdi
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Myrto Moutafi
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Ioannis Kotsantis
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Lampis C. Stavrinou
- Department of Neurosurgery and Neurotraumatology, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Amanda Psyrri
- Department of Medical Oncology, Second Department of Internal Medicine, “Attikon” University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|