1
|
Kulkarni T, Banik S, Mukhopadhyay D, Babiker H, Bhattacharya S. Tumor-Treating Fields Alter Nanomechanical Properties of Pancreatic Ductal Adenocarcinoma Cells Co-Cultured with Extracellular Matrix. J Funct Biomater 2025; 16:160. [PMID: 40422825 DOI: 10.3390/jfb16050160] [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: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
Tumor-Treating Fields (TTFields), a novel therapeutic avenue, is approved for therapy in Glioblastoma multiforme, malignant pleural mesothelioma, and metastatic non-small cell lung cancer (NSCLC). In pancreatic ductal adenocarcinoma (PDAC), several clinical trials are underway to improve outcomes, yet a significant knowledge gap prevails involving the cell-extracellular matrix (ECM) crosstalk. Herein, we hypothesized that treatment with TTFields influence this crosstalk, which is reflected by the dynamic alteration in nanomechanical properties (NMPs) of cells and the ECM in a co-culture system. We employed an ECM gel comprising collagen, fibronectin, and laminin mixed in 100:1:1 stoichiometry to co-culture of Panc1 and AsPC1 individually. This ECM mixture mimics the in vivo tumor microenvironment closely when compared to the individual ECM components studied before. A comprehensive frequency-dependent study revealed the optimal TTFields frequency to be 150 kHz. We also observed that irrespective of the ECM's presence, TTFields increase cell membrane stiffness and decrease deformation several-folds in both Panc1 and AsPC1 cells at both 48 h and 72 h. Although adhesion for AsPC1 decreased at 48 h, at 72 h it was observed to increase irrespective of ECM's presence. Moreover, it significantly alters the NMPs of ECM gels when co-cultured with PDAC cell lines. However, AsPC1 cells were observed to be more detrimental to these changes. Lastly, we attribute the stiffness changes in Panc1 cells to the membrane F-actin reorganization in the presence of TTFields. This study paves a path to study complex PDAC TME as well as the effect of various chemotherapeutic agents on such TME with TTFields in the future.
Collapse
Affiliation(s)
- Tanmay Kulkarni
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Sreya Banik
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Hani Babiker
- Department of Medicine, Division of Hematology and Oncology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
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
|
5
|
Klein-Goldberg A, Voloshin T, Zemer Tov E, Paz R, Somri-Gannam L, Volodin A, Koren L, Lifshitz L, Meir A, Shabtay-Orbach A, Blatt R, Cahal S, Tempel-Brami C, Wainer-Katsir K, Kan T, Koltun B, Brant B, Barsheshet Y, Haber A, Giladi M, Weinberg U, Palti Y. Role of the PI3K/AKT signaling pathway in the cellular response to Tumor Treating Fields (TTFields). Cell Death Dis 2025; 16:210. [PMID: 40148314 PMCID: PMC11950169 DOI: 10.1038/s41419-025-07546-8] [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] [Received: 08/14/2024] [Revised: 02/18/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Tumor Treating Fields (TTFields) are electric fields that induce cancer cell death. Genomic analysis of glioblastoma tumors resected from TTFields-treated patients suggested a potential link between a reduced or absent response to TTFields and activating mutations in the phosphatidylinositol 3-kinase (PI3K) p110α subunit (PIK3CA). Our study aimed to investigate the role of the PI3K/AKT pathway in the response to TTFields. We tested changes in signaling pathways in control versus TTFields-treated U-87 MG glioblastoma, A2780 ovarian carcinoma, and H1299 non-small cell lung cancer (NSCLC) cells using the Luminex multiplex assay, validated by western blot analysis and inhibition assays. We also performed in vivo validation using immunohistochemistry on tumor sections from animals bearing orthotopic N1-S1 hepatocellular, MOSE-L ovarian, or LL/2 lung tumors that were treated with TTFields or sham. Finally, we examined the efficacy of concomitant treatment with TTFields and PI3K inhibitors in cell lines and mouse models. Our findings elucidate the mechanisms driving PI3K/AKT activation following TTFields treatment, revealing that the AKT signaling amplitude increases over time and is influenced by cell-surface and cell-cell interactions. Specifically, focal adhesion kinase (FAK) and N-cadherin were found to promote AKT phosphorylation, activating cell survival pathways. Furthermore, our investigation revealed that pharmacological inhibition of PI3K sensitized cancer cells to TTFields, both in vitro and in vivo. Our research suggests that the PI3K/AKT pathway is involved in cancer cell response to TTFields, and that inhibition of this pathway may serve as a potential therapeutic target for sensitizing cancer cells to TTFields.
Collapse
|
6
|
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
|
7
|
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
|
8
|
Kalita S, Danovich D, Shaik S. Origins of the Superiority of Oscillating Electric Fields for Disrupting Senile Plaques: Insights from the 7-Residue Fragment and the Full-length Aβ-42 Peptide. J Am Chem Soc 2025; 147:2626-2641. [PMID: 39772489 PMCID: PMC11760182 DOI: 10.1021/jacs.4c14791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/01/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Our recent molecular dynamics simulations of decomposing Alzheimer's disease plaques, under oscillating- and static external electric fields (Os-EEFs and St-EEFs), revealed the superiority of Os-EEF for decomposing plaques consisting of the 7-residue peptide segment. This conclusion is now reinforced by studying the dimers of the short peptides and trimers of the full-length Aβ-42 peptide. Thus, the dispersed peptides obtained following St-EEF applications reformed the plaques once the St-EEF subsided. In contrast, plaques originating from the application of Os-EEF remained dispersed for long time scales. The present study provides insights into these results by modeling the decomposition modes that transpire under both field types. Additionally, this study provides insights into the frequency effects on the decomposition processes within the THz-MHz regions. The simulation shows that the Os-EEF in the lower frequency range (≤GHz) decomposes the plaque on a time scale of ∼50 ns, whereas the higher frequency Os-EEFs (≥THz) are less effective. As such, Os-EEFs with moderate-to-low frequencies (≤GHz) lead to an "explosion," whereby the peptides fly distantly apart and inhibit plaque reformation. By contrast, St-EEFs form parallel peptide pairs, which are stabilized by the EEF due to the large dipole moment of the ensemble. Thus, St-EEF applications lead to sluggish and reversible plaque decomposition processes. We further conclude that the Os-EEF impact is maximal for short pulses, which prevents the EEF propensity to arrange the peptides in parallel pairs. The superiority of the Os-EEF over the St-EEF is maintained irrespective of the peptides' length. A model is formulated that predicts the dependence of the decomposition time scale on the EEF.
Collapse
Affiliation(s)
- Surajit Kalita
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 9190401, Israel
| | - David Danovich
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 9190401, Israel
| | - Sason Shaik
- Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 9190401, Israel
| |
Collapse
|
9
|
Smothers AR, Beasley ME, Warren HS, Kegel OG, Edenfield WJ, O’Connell JJ, Booth BW. Tumor-treating fields and concurrent cisplatin: an in vitro demonstration of efficacy in triple-negative breast cancer. Am J Cancer Res 2025; 15:322-330. [PMID: 39949927 PMCID: PMC11815381 DOI: 10.62347/lxjh5896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Chemotherapy is commonly used to treat patients with triple-negative breast cancer. Combinations of platinum-based chemotherapies have demonstrated higher rates of pathologic complete responses of triple-negative breast cancer compared to combinations without platinum-based chemotherapies. However, there is a significant increase in general toxicity with the addition of platinum-based regimens. Some groups have investigated using tumor-treating fields as an alternative treatment method for triple-negative breast cancer, and chemosensitization by tumor-treating fields has been observed in vitro. With the goal of minimizing the toxicities associated with platinum-based chemotherapy, we investigated anti-mitotic effects of concurrent tumor-treating fields and cisplatin treatment to show that the addition of tumor-treating fields further inhibited cell growth in triple-negative breast cancer. We show that combining cisplatin with tumor-treating fields induces higher levels of apoptosis within triple-negative breast cancer cells as opposed to normal epithelial cells and the combination has a more immediate mechanism of cell death than either monotherapy. We also demonstrate that, when combined with tumor-treating fields, a lower dose of chemotherapy can be used to achieve the same efficacy of triple-negative breast cancer cell death as higher doses of individual therapy.
Collapse
Affiliation(s)
- Austin R Smothers
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson UniversityClemson, SC, USA
- Department of Bioengineering, Clemson UniversityClemson, SC, USA
| | - Mya E Beasley
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson UniversityClemson, SC, USA
- Department of Bioengineering, Clemson UniversityClemson, SC, USA
| | - Hunter S Warren
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson UniversityClemson, SC, USA
| | - Olivia G Kegel
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson UniversityClemson, SC, USA
| | - W Jeffery Edenfield
- Prisma Health Cancer Institute, Prisma HealthGreenville, SC, USA
- Institute for Translational Oncological Research, Prisma HealthGreenville, SC, USA
| | - John J O’Connell
- University of South Carolina School of Medicine-GreenvilleGreenville, SC, USA
- Prisma Health Cancer Institute, Prisma HealthGreenville, SC, USA
- Clemson University School of Health ResearchClemson, SC, USA
| | - Brian W Booth
- Department of Bioengineering, Clemson UniversityClemson, SC, USA
- Clemson University School of Health ResearchClemson, SC, USA
| |
Collapse
|
10
|
Xiao T, Zheng H, Zu K, Yue Y, Wang Y. Tumor-treating fields in cancer therapy: advances of cellular and molecular mechanisms. Clin Transl Oncol 2025; 27:1-14. [PMID: 38884919 DOI: 10.1007/s12094-024-03551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Tumor-Treating Fields (TTFields) use intermediate-frequency and low-intensity electric fields to inhibit tumor cells. However, their mechanisms are still not well understood. This article reviews their key antitumor mechanisms at the cellular and molecular levels, including inhibition of proliferation, induction of death, disturbance of migration, and activation of the immune system. The multifaceted biological effects in combination with other cancer treatments are also summarized. The deep insight into their mechanism will help develop more potential antitumor treatments.
Collapse
Affiliation(s)
- Tong Xiao
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Hao Zheng
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Kaiyang Zu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Youjia Yue
- School of Biomedical Engineeringg, Capital Medical University, Beijing, 100069, China
| | - Ying Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
11
|
Lok E, Chang B, Vega R, Haack M, Wong ET. Effects of craniectomy defect on tumor-treating fields. Neurooncol Adv 2025; 7:vdaf045. [PMID: 40376683 PMCID: PMC12080538 DOI: 10.1093/noajnl/vdaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025] Open
Abstract
Background Tumor-treating fields (TTFields) are alternating electric fields approved for the treatment of glioblastoma. They must penetrate through the skull to reach the gross tumor volume (GTV) in the brain. Since the skull is an attenuator of electric fields, removal of a section of cortical bone by craniectomy may facilitate the delivery of TTFields into the GTV. Methods We identified a glioblastoma patient who underwent craniectomy for evacuation of a subdural empyema. The patient subsequently received standard adjuvant treatment with TTFields plus temozolomide without replacement of the skull defect. Post-acquisition magnetic resonance imaging datasets were obtained from this index patient and 2 others for virtual craniectomy analysis. After anatomic delineation, a 3-dimensional finite element mesh was generated and then solved for the distribution of applied electric fields, rate of energy deposition, and current density at the GTV. Results The geometry of craniectomy defect alone, with or without burr holes, did not alter TTFields delivery to GTV. Biomaterials filling the defect could significantly influence electric field penetration, particularly when they are highly conductive at 10 S/m or 7.76 × 106 S/m as in tantalum. The ratio of GTV relative to defect size also enhanced or attenuated TTFields coverage when the GTV was expanded or eroded, respectively. Conclusions Craniectomy, biomaterials filling the defect, and the ratio of GTV relative to defect size may interact in a combinatorial fashion in modulating TTFields penetration into the brain. These findings are clinically relevant for personalized TTFields treatment.
Collapse
Affiliation(s)
- Edwin Lok
- Division of Hematology/Oncology, Department of Medicine, Brown University Health & Rhode Island Hospital, Providence, Rhode Island, USA
- Brain Tumor Center & Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Bryant Chang
- Division of Neurosurgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Rafael Vega
- Division of Neurosurgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Monika Haack
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Eric T Wong
- Department of Neurology, Medicine, Neurosurgery & Radiation Oncology, Brown University Health & Rhode Island Hospital, Providence, Rhode Island, USA
- Division of Hematology/Oncology, Department of Medicine, Brown University Health & Rhode Island Hospital, Providence, Rhode Island, USA
- Brain Tumor Center & Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Zhang X, Yan Z, Huang L, Yu X, Huang R. A new tumor-treating device OM-100 with low-frequency magnetic fields inhibits proliferation and metastasis in liver cancer. BMC Cancer 2024; 24:1383. [PMID: 39528972 PMCID: PMC11552396 DOI: 10.1186/s12885-024-13121-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This study aims to investigate a novel instrument OM-100 with low-frequency magnetic fields (LFMFs) for its potential applicability in the treatment of liver cancer. METHODS Liver cancer cell lines (HepG2 and Huh7) and normal liver cell line THLE-2 were exposed to OM-100 at LFMFs of 0, 10, 25, 50, and 100 kHz for 2 h in the morning, noon, and evening, respectively. The effects of LFMF on cell viability, apoptosis, migration, and invasion capabilities were examined. Additionally, impacts of LFMF on ROS production was assessed. In vivo studies were conducted to examine the safety profile of OM-100 and its effects on tumor growth. RESULTS In vitro, OM-100 reduced the viability of liver cancer cells, increased cell apoptosis, and inhibited cell migration and invasion abilities in a frequency-dependent manner (P < 0.05). In vivo, OM-100 significantly slowed down tumor growth and promoted apoptosis in liver tumors (P < 0.05). Moreover, OM-100 rarely affected the viability of normal liver cells, as well as the health of normal mice. Finally, we further found that OM-100 significantly increased the production of ROS in liver cancer cells (P < 0.05), a key factor in inducing autophagy, which is very important for the progression of liver cancer. CONCLUSION Our findings reveal the safety of OM-100 and its frequency at 100 kHz significantly inhibits liver cancer progression.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Zhaoxian Yan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lifa Huang
- Department of Neurosurgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, China
| | - Xinyan Yu
- Department of Medical, Ci Xing Technology Co., Ltd, No. 100, Changhe Road, Hangzhou, 310051, China
| | - Rui Huang
- Department of Medical, Ci Xing Technology Co., Ltd, No. 100, Changhe Road, Hangzhou, 310051, China.
| |
Collapse
|
13
|
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
|
14
|
Pan J, Eskandar T, Ahmed Z, Agrawal DK. Biophysical and Biological Mechanisms of Tumor Treating Fields in Glioblastoma. JOURNAL OF CANCER SCIENCE AND CLINICAL THERAPEUTICS 2024; 8:265-270. [PMID: 39364266 PMCID: PMC11448370 DOI: 10.26502/jcsct.5079249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Glioblastoma (GBM) is one of the most aggressive forms of brain cancer that presents with a median survival rate of 14-30 months and along with a discouraging five-year survival rate of 4-5%. Standard treatment of newly diagnosed GBM, also known as the Stupp protocol, includes a maximally safe surgical resection followed by radiation and chemotherapy. Despite these treatment regimens, recurrence is almost inevitable, emphasizing the need for new therapies to combat the aggressive nature of GBMs. Tumor Treating Fields (TTFs) are a relatively new application to the treatment of GBMs, and results have been promising with both progression-free survival and overall survival when TTFs have been used in combination with temozolomide. This article critically reviews the biophysical and biological mechanisms of TTFs, their clinical efficacy, and discusses the results in clinical trials, including EF-11 and EF-14. Both trials have demonstrated that TTFs can enhance progression free survival and overall survival without compromising quality of life or causing severe adverse effects. Despite the high cost associated with TTFs and the need for further analysis to determine the most effective ways to integrate TTFs into GBM treatments, TTFs represent a significant advancement in GBM therapy and offer hope for improved patient prognosis.
Collapse
Affiliation(s)
- Jeremy Pan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Tony Eskandar
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Zubair Ahmed
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| |
Collapse
|
15
|
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
|
16
|
Song B, Wang X, Qin L, Hussain S, Liang W. Brain gliomas: Diagnostic and therapeutic issues and the prospects of drug-targeted nano-delivery technology. Pharmacol Res 2024; 206:107308. [PMID: 39019336 DOI: 10.1016/j.phrs.2024.107308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Glioma is the most common intracranial malignant tumor, with severe difficulty in treatment and a low patient survival rate. Due to the heterogeneity and invasiveness of tumors, lack of personalized clinical treatment design, and physiological barriers, it is often difficult to accurately distinguish gliomas, which dramatically affects the subsequent diagnosis, imaging treatment, and prognosis. Fortunately, nano-delivery systems have demonstrated unprecedented capabilities in diagnosing and treating gliomas in recent years. They have been modified and surface modified to efficiently traverse BBB/BBTB, target lesion sites, and intelligently release therapeutic or contrast agents, thereby achieving precise imaging and treatment. In this review, we focus on nano-delivery systems. Firstly, we provide an overview of the standard and emerging diagnostic and treatment technologies for glioma in clinical practice. After induction and analysis, we focus on summarizing the delivery methods of drug delivery systems, the design of nanoparticles, and their new advances in glioma imaging and treatment in recent years. Finally, we discussed the prospects and potential challenges of drug-delivery systems in diagnosing and treating glioma.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Shehbaz Hussain
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
17
|
Dieper A, Scheidegger S, Füchslin RM, Veltsista PD, Stein U, Weyland M, Gerster D, Beck M, Bengtsson O, Zips D, Ghadjar P. Literature review: potential non-thermal molecular effects of external radiofrequency electromagnetic fields on cancer. Int J Hyperthermia 2024; 41:2379992. [PMID: 39019469 DOI: 10.1080/02656736.2024.2379992] [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: 02/16/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
INTRODUCTION There is an ongoing scientific discussion, that anti-cancer effects induced by radiofrequency (RF)-hyperthermia might not be solely attributable to subsequent temperature elevations at the tumor site but also to non-temperature-induced effects. The exact molecular mechanisms behind said potential non-thermal RF effects remain largely elusive, however, limiting their therapeutical targetability. OBJECTIVE Therefore, we aim to provide an overview of the current literature on potential non-temperature-induced molecular effects within cancer cells in response to RF-electromagnetic fields (RF-EMF). MATERIAL AND METHODS This literature review was conducted following the PRISMA guidelines. For this purpose, a MeSH-term-defined literature search on MEDLINE (PubMed) and Scopus (Elsevier) was conducted on March 23rd, 2024. Essential criteria herein included the continuous wave RF-EMF nature (3 kHz - 300 GHz) of the source, the securing of temperature-controlled circumstances within the trials, and the preclinical nature of the trials. RESULTS Analysis of the data processed in this review suggests that RF-EMF radiation of various frequencies seems to be able to induce significant non-temperature-induced anti-cancer effects. These effects span from mitotic arrest and growth inhibition to cancer cell death in the form of autophagy and apoptosis and appear to be mostly exclusive to cancer cells. Several cellular mechanisms were identified through which RF-EMF radiation potentially imposes its anti-cancer effects. Among those, by reviewing the included publications, we identified RF-EMF-induced ion channel activation, altered gene expression, altered membrane potentials, membrane oscillations, and blebbing, as well as changes in cytoskeletal structure and cell morphology. CONCLUSION The existent literature points toward a yet untapped therapeutic potential of RF-EMF treatment, which might aid in damaging cancer cells through bio-electrical and electro-mechanical molecular mechanisms while minimizing adverse effects on healthy tissue cells. Further research is imperative to definitively confirm non-thermal EMF effects as well as to determine optimal cancer-type-specific RF-EMF frequencies, field intensities, and exposure intervals.
Collapse
Affiliation(s)
- Anna Dieper
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stephan Scheidegger
- Institute for Applied Mathematics and Physics, Zurich University of Applied Sciences, Winterthur, Switzerland
| | - Rudolf M Füchslin
- Institute for Applied Mathematics and Physics, Zurich University of Applied Sciences, Winterthur, Switzerland
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum (MDC), Berlin, Germany
| | - Paraskevi D Veltsista
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum (MDC), Berlin, Germany
| | - Mathias Weyland
- Institute for Applied Mathematics and Physics, Zurich University of Applied Sciences, Winterthur, Switzerland
| | - Dominik Gerster
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Beck
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Olof Bengtsson
- Ferdinand-Braun-Institut (FBH), Leibnitz-Institut für Höchstfrequenztechnik, Berlin, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
18
|
Tian J, Mallinger JC, Shi P, Ling D, Deleyrolle LP, Lin M, Khoshbouei H, Sarkisian MR. Aurora kinase A inhibition plus Tumor Treating Fields suppress glioma cell proliferation in a cilium-independent manner. Transl Oncol 2024; 45:101956. [PMID: 38640786 PMCID: PMC11053227 DOI: 10.1016/j.tranon.2024.101956] [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: 12/04/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/21/2024] Open
Abstract
Tumor Treating Fields (TTFields) extend the survival of glioblastoma (GBM) patients by interfering with a broad range of tumor cellular processes. Among these, TTFields disrupt primary cilia stability on GBM cells. Here we asked if concomitant treatment of TTFields with other agents that interfere with GBM ciliogenesis further suppress GBM cell proliferation in vitro. Aurora kinase A (AURKA) promotes both cilia disassembly and GBM growth. Inhibitors of AURKA, such as Alisertib, inhibit cilia disassembly and increase ciliary frequency in various cell types. However, we found that Alisertib treatment significantly reduced GBM cilia frequency in gliomaspheres across multiple patient derived cell lines, and in patient biopsies treated ex vivo. This effect appeared glioma cell-specific as it did not reduce normal neuronal or glial cilia frequencies. Alisertib-mediated depletion of glioma cilia appears specific to AURKA and not AURKB inhibition, and attributable in part to autophagy pathway activation. Treatment of two different GBM patient-derived cell lines with TTFields and Alisertib resulted in a significant reduction in cell proliferation compared to either treatment alone. However, this effect was not cilia-dependent as the combined treatment reduced proliferation in cilia-depleted cell lines lacking, ARL13B, or U87MG cells which are naturally devoid of ARL13B+ cilia. Thus, Alisertib-mediated effects on glioma cilia may be a useful biomarker of drug efficacy within tumor tissue. Considering Alisertib can cross the blood brain barrier and inhibit intracranial growth, our data warrant future studies to explore whether concomitant Alisertib and TTFields exposure prolongs survival of brain tumor-bearing animals in vivo.
Collapse
Affiliation(s)
- Jia Tian
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA; Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Julianne C Mallinger
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA; Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Ping Shi
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA; Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Dahao Ling
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA; Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Loic P Deleyrolle
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Min Lin
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Matthew R Sarkisian
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA; Preston A. Wells Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
19
|
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
|
20
|
Xing Y, Yasinjan F, Cui J, Peng Y, He M, Liu W, Hong X. Advancements and current trends in tumor treating fields: a scientometric analysis. Int J Surg 2024; 110:2978-2991. [PMID: 38349201 PMCID: PMC11093503 DOI: 10.1097/js9.0000000000001151] [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: 10/06/2023] [Accepted: 01/26/2024] [Indexed: 05/16/2024]
Abstract
Tumor treating fields (TTFields) therapy is a novel and effective noninvasive cancer therapy, and it has been approved by FDA in the treatment of recurrent and newly diagnosed glioblastoma, and malignant pleural mesothelioma. Moreover, TTFields therapy has been widely studied in both clinical trials and preclinical studies in recent years. Based on its high efficacy, research on TTFields therapy has been a hot topic. Thus, the authors made this scientometric analysis of TTfields to reveal the scientometric distributions such as annual publications and citations, countries and institutions, authors, journals, references, and more importantly, research status and hot topics of the field. In recent years, publication numbers have been stable at high values, and citation numbers have been increasing greatly. The United States and Israel were the top two countries with the highest publication numbers, followed by Germany and Switzerland. Scientometric analyses of keywords indicated that clinical applications and antitumor mechanisms are probably the two main parts of current research on TTfields. Most clinical trials of TTfields focus on the treatment of glioblastoma. And a variety of other cancers such as lung cancer especially nonsmall cell lung cancer, hepatic cancer, other brain tumors, etc. have also been studied in both clinical trials and preclinical studies.
Collapse
Affiliation(s)
- Yang Xing
- Department of Neurosurgery, The First Hospital of Jilin University
| | - Feroza Yasinjan
- Department of Neurosurgery, The First Hospital of Jilin University
| | - Jiayue Cui
- Department of Histology and Embryology, College of Basic Medical Sciences
| | - Yizhao Peng
- Department of Neurosurgery, The First Hospital of Jilin University
| | - Minghua He
- College of Computer Science and Technology, Jilin University, Changchun, People’s Republic of China
| | - Wenhui Liu
- Department of Histology and Embryology, College of Basic Medical Sciences
| | - Xinyu Hong
- Department of Neurosurgery, The First Hospital of Jilin University
| |
Collapse
|
21
|
Lan J, Liu Y, Chen J, Liu H, Feng Y, Liu J, Chen L. Advanced tumor electric fields therapy: A review of innovative research and development and prospect of application in glioblastoma. CNS Neurosci Ther 2024; 30:e14720. [PMID: 38715344 PMCID: PMC11077002 DOI: 10.1111/cns.14720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive malignant tumor with a high mortality rate and is the most prevalent primary intracranial tumor that remains incurable. The current standard treatment, which involves surgery along with concurrent radiotherapy and chemotherapy, only yields a survival time of 14-16 months. However, the introduction of tumor electric fields therapy (TEFT) has provided a glimmer of hope for patients with newly diagnosed and recurrent GBM, as it has been shown to extend the median survival time to 20 months. The combination of TEFT and other advanced therapies is a promising trend in the field of GBM, facilitated by advancements in medical technology. AIMS In this review, we provide a concise overview of the mechanism and efficacy of TEFT. In addition, we mainly discussed the innovation of TEFT and our proposed blueprint for TEFT implementation. CONCLUSION Tumor electric fields therapy is an effective and highly promising treatment modality for GBM. The full therapeutic potential of TEFT can be exploited by combined with other innovative technologies and treatments.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| | - Yuyang Liu
- Medical School of Chinese PLABeijingChina
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Junyi Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Hongyu Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryHainan Hospital of Chinese PLA General HospitalHainanChina
| | - Yaping Feng
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jialin Liu
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Ling Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| |
Collapse
|
22
|
秦 丽, 谢 旭, 王 敏, 马 明, 潘 赟, 陈 光, 张 韶. [Simulation model of tumor-treating fields]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2024; 41:360-367. [PMID: 38686418 PMCID: PMC11058494 DOI: 10.7507/1001-5515.202306074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 01/04/2024] [Indexed: 05/02/2024]
Abstract
Tumor-treating fields (TTFields) is a novel treatment modality for malignant solid tumors, often employing electric field simulations to analyze the distribution of electric fields on the tumor under different parameters of TTFields. Due to the present difficulties and high costs associated with reproducing or implementing the simulation model construction techniques, this study used readily available open-source software tools to construct a highly accurate, easily implementable finite element simulation model for TTFields. The accuracy of the model is at a level of 1 mm 3. Using this simulation model, the study carried out analyses of different factors, such as tissue electrical parameters and electrode configurations. The results show that factors influncing the distribution of the internal electric field of the tumor include changes in scalp and skull conductivity (with a maximum variation of 21.0% in the treatment field of the tumor), changes in tumor conductivity (with a maximum variation of 157.8% in the treatment field of the tumor), and different electrode positions and combinations (with a maximum variation of 74.2% in the treatment field of the tumor). In summary, the results of this study validate the feasibility and effectiveness of the proposed modeling method, which can provide an important reference for future simulation analyses of TTFields and clinical applications.
Collapse
Affiliation(s)
- 丽平 秦
- 浙江省医疗器械检验研究院(杭州 310018)Zhejiang Institute of Medical Device Testing, Hangzhou 310018, P. R. China
| | - 旭 谢
- 浙江省医疗器械检验研究院(杭州 310018)Zhejiang Institute of Medical Device Testing, Hangzhou 310018, P. R. China
- 浙江大学 求是高等研究院 浙江大学生物医学工程教育部重点实验室(杭州 310027)Qiushi Academy for Advanced Studies, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou 310027, P. R. China
| | - 敏敏 王
- 浙江省医疗器械检验研究院(杭州 310018)Zhejiang Institute of Medical Device Testing, Hangzhou 310018, P. R. China
| | - 明伟 马
- 浙江省医疗器械检验研究院(杭州 310018)Zhejiang Institute of Medical Device Testing, Hangzhou 310018, P. R. China
| | - 赟 潘
- 浙江省医疗器械检验研究院(杭州 310018)Zhejiang Institute of Medical Device Testing, Hangzhou 310018, P. R. China
| | - 光弟 陈
- 浙江省医疗器械检验研究院(杭州 310018)Zhejiang Institute of Medical Device Testing, Hangzhou 310018, P. R. China
| | - 韶岷 张
- 浙江省医疗器械检验研究院(杭州 310018)Zhejiang Institute of Medical Device Testing, Hangzhou 310018, P. R. China
| |
Collapse
|
23
|
Nasori N, Firdhaus M, Farahdina U, Khamimatul Ula R. Optimizing tumor treating fields for blood cancer therapy: Analysis of electric field distribution and dose density. Biophys Physicobiol 2024; 21:e210013. [PMID: 39206129 PMCID: PMC11347819 DOI: 10.2142/biophysico.bppb-v21.0013] [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: 09/29/2023] [Accepted: 04/16/2024] [Indexed: 09/04/2024] Open
Abstract
Blood cancer is a condition in which white blood cells grow uncontrollably. Tumor treating fields (TTF) are a modality of cancer treatment that utilizes electric fields to target malignant cells. To optimize the efficacy of TTF, it is necessary to investigate the distribution of electric field through varying electrode configurations and input parameters. This allows for enhancement of electric field intensity in targeted areas while minimizing intensity in sensitive areas. Analysis of electric field distribution was conducted through simulation of brachial models with varying electrode configurations and input parameters, utilizing the COMSOL Multiphysics 5.4 software. Additionally, investigations were carried out to assess tissue dose density. The dose density value at primary target for all electrode configurations and input parameters do not exceed the threshold value (770 W/m3), whereas the electric field value at the primary target satisfied the threshold value (100 V/m) on the system that used 4 plate-shaped electrodes and arm contour-shaped electrodes with an input voltage of 20 V, and at the input voltage 15 V, only 4 arm contour-shaped electrodes that satisfied the threshold value. An increase in input voltage, electrodes addition, and electrodes adjustment to skin contour shape result in an enhancement of electric field distribution and average electric field value at primary targets.
Collapse
Affiliation(s)
- Nasori Nasori
- Laboratory of Medical Physics and Biophysics, Department of Physics, Faculty Science and Data Analytic, Institute Teknologi Sepuluh Nopember, Surabaya 60111, Indonesia
| | - Miftakhul Firdhaus
- Laboratory of Medical Physics and Biophysics, Department of Physics, Faculty Science and Data Analytic, Institute Teknologi Sepuluh Nopember, Surabaya 60111, Indonesia
| | - Ulya Farahdina
- Laboratory of Medical Physics and Biophysics, Department of Physics, Faculty Science and Data Analytic, Institute Teknologi Sepuluh Nopember, Surabaya 60111, Indonesia
| | - Rini Khamimatul Ula
- Research Center Of Electronics, National Research and Innovation Agency of Indonesia (BRIN), Jakarta, 10340, Indonesia and Nusa Putra University, Sukabumi, Indonesia
| |
Collapse
|
24
|
Yu A, Zeng J, Yu J, Cao S, Li A. Theory and application of TTFields in newly diagnosed glioblastoma. CNS Neurosci Ther 2024; 30:e14563. [PMID: 38481068 PMCID: PMC10938032 DOI: 10.1111/cns.14563] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Glioblastoma is the most common primary malignant brain tumor in adults. TTFields is a therapy that use intermediate-frequency and low-intensity alternating electric fields to treat tumors. For patients with ndGBM, the addition of TTFields after the concurrent chemoradiotherapy phase of the Stupp regimen can improve prognosis. However, TTFields still has the potential to further prolong the survival of ndGBM patients. AIM By summarizing the mechanism and application status of TTFields in the treatment of ndGBM, the application prospect of TTFields in ndbm treatment is prospected. METHODS We review the recent literature and included 76 articles to summarize the mechanism of TTfields in the treatment of ndGBM. The current clinical application status and potential health benefits of TTFields in the treatment of ndGBM are also discussed. RESULTS TTFields can interfere with tumor cell mitosis, lead to tumor cell apoptosis and increased autophagy, hinder DNA damage repair, induce ICD, activate tumor immune microenvironment, reduce cancer cell metastasis and invasion, and increase BBB permeability. TTFields combines with chemoradiotherapy has made progress, its optimal application time is being explored and the problems that need to be considered when retaining the electrode patches for radiotherapy are further discussed. TTFields shows potential in combination with immunotherapy, antimitotic agents, and PARP inhibitors, as well as in patients with subtentorial gliomas. CONCLUSION This review summarizes mechanisms of TTFields in the treatment of ndGBM, and describes the current clinical application of TTFields in ndGBM. Through the understanding of its principle and application status, we believe that TTFields still has the potential to further prolong the survival of ndGBM patients. Thus,research is still needed to explore new ways to combine TTFields with other therapies and optimize the use of TTFields to realize its full potential in ndGBM patients.
Collapse
Affiliation(s)
- Ao Yu
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical UniversityCancer Hospital of Dalian University of TechnologyShenyangChina
- School of GraduateChina Medical UniversityShenyangChina
| | - Juan Zeng
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Jinhui Yu
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical UniversityCancer Hospital of Dalian University of TechnologyShenyangChina
- School of GraduateChina Medical UniversityShenyangChina
| | - Shuo Cao
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Ailin Li
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical UniversityCancer Hospital of Dalian University of TechnologyShenyangChina
| |
Collapse
|
25
|
Ravin R, Cai TX, Li A, Briceno N, Pursley RH, Garmendia-Cedillos M, Pohida T, Wang H, Zhuang Z, Cui J, Morgan NY, Williamson NH, Gilbert MR, Basser PJ. "Tumor Treating Fields" delivered via electromagnetic induction have varied effects across glioma cell lines and electric field amplitudes. Am J Cancer Res 2024; 14:562-584. [PMID: 38455403 PMCID: PMC10915321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/15/2023] [Indexed: 03/09/2024] Open
Abstract
Previous studies reported that alternating electric fields (EFs) in the intermediate frequency (100-300 kHz) and low intensity (1-3 V/cm) regime - termed "Tumor Treating Fields" (TTFields) - have a specific, anti-proliferative effect on glioblastoma multiforme (GBM) cells. However, the mechanism(s) of action remain(s) incompletely understood, hindering the clinical adoption of treatments based on TTFields. To advance the study of such treatment in vitro, we developed an inductive device to deliver EFs to cell cultures which improves thermal and osmolar regulation compared to prior devices. Using this inductive device, we applied continuous, 200 kHz electromagnetic fields (EMFs) with a radial EF amplitude profile spanning 0-6.5 V/cm to cultures of primary rat astrocytes and several human GBM cell lines - U87, U118, GSC827, and GSC923 - for a duration of 72 hours. Cell density was assessed via segmented pixel densities from GFP expression (U87, U118) or from staining (astrocytes, GSC827, GSC923). Further RNA-Seq analyses were performed on GSC827 and GSC923 cells. Treated cultures of all cell lines exhibited little to no change in proliferation at lower EF amplitudes (0-3 V/cm). At higher amplitudes (> 4 V/cm), different effects were observed. Apparent cell densities increased (U87), decreased (GSC827, GSC923), or showed little change (U118, astrocytes). RNA-Seq analyses on treated and untreated GSC827 and GSC923 cells revealed differentially expressed gene sets of interest, such as those related to cell cycle control. Up- and down-regulation, however, was not consistent across cell lines nor EF amplitudes. Our results indicate no consistent, anti-proliferative effect of 200 kHz EMFs across GBM cell lines and thus contradict previous in vitro findings. Rather, effects varied across different cell lines and EF amplitude regimes, highlighting the need to assess the effect(s) of TTFields and similar treatments on a per cell line basis.
Collapse
Affiliation(s)
- Rea Ravin
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIHBethesda, Maryland, USA
- Celoptics, Inc.Rockville, Maryland, USA
| | - Teddy X Cai
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIHBethesda, Maryland, USA
- The Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, Oxford UniversityOxfordshire, UK
| | - Aiguo Li
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIHBethesda, Maryland, USA
| | - Nicole Briceno
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIHBethesda, Maryland, USA
| | - Randall H Pursley
- Instrumentation Development and Engineering Applications Section, National Institute of Biomedical Imaging and Bioengineering, NIHBethesda, Maryland, USA
| | - Marcial Garmendia-Cedillos
- Instrumentation Development and Engineering Applications Section, National Institute of Biomedical Imaging and Bioengineering, NIHBethesda, Maryland, USA
| | - Tom Pohida
- Instrumentation Development and Engineering Applications Section, National Institute of Biomedical Imaging and Bioengineering, NIHBethesda, Maryland, USA
| | - Herui Wang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIHBethesda, Maryland, USA
| | - Zhengping Zhuang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIHBethesda, Maryland, USA
| | - Jing Cui
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIHBethesda, Maryland, USA
| | - Nicole Y Morgan
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences, National Institute of Biomedical Imaging and Bioengineering, NIHBethesda, Maryland, USA
| | - Nathan H Williamson
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIHBethesda, Maryland, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIHBethesda, Maryland, USA
| | - Peter J Basser
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIHBethesda, Maryland, USA
| |
Collapse
|
26
|
Ni X, Yu X, Ye Q, Su X, Shen S. Desflurane improves electrical activity of neurons and alleviates oxygen-glucose deprivation-induced neuronal injury by activating the Kcna1-dependent Kv1.1 channel. Exp Brain Res 2024; 242:477-490. [PMID: 38184806 DOI: 10.1007/s00221-023-06764-w] [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: 09/12/2023] [Accepted: 12/11/2023] [Indexed: 01/08/2024]
Abstract
Several volatile anesthetics have presented neuroprotective functions in ischemic injury. This study investigates the effect of desflurane (Des) on neurons following oxygen-glucose deprivation (OGD) challenge and explores the underpinning mechanism. Mouse neurons HT22 were subjected to OGD, which significantly reduced cell viability, increased lactate dehydrogenase release, and promoted cell apoptosis. In addition, the OGD condition increased oxidative stress in HT22 cells, as manifested by increased ROS and MDA contents, decreased SOD activity and GSH/GSSG ratio, and reduced nuclear protein level of Nrf2. Notably, the oxidative stress and neuronal apoptosis were substantially blocked by Des treatment. Bioinformatics suggested potassium voltage-gated channel subfamily A member 1 (Kcna1) as a target of Des. Indeed, the Kcna1 expression in HT22 cells was decreased by OGD but restored by Des treatment. Artificial knockdown of Kcna1 negated the neuroprotective effects of Des. By upregulating Kcna1, Des activated the Kv1.1 channel, therefore enhancing K+ currents and inducing neuronal repolarization. Pharmacological inhibition of the Kv1.1 channel reversed the protective effects of Des against OGD-induced injury. Collectively, this study demonstrates that Des improves electrical activity of neurons and alleviates OGD-induced neuronal injury by activating the Kcna1-dependent Kv1.1 channel.
Collapse
Affiliation(s)
- Xiaolei Ni
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, No. 120, Suzhi Road, Sucheng District, Suqian, 223800, Jiangsu, People's Republic of China
| | - Xiaoyan Yu
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, No. 120, Suzhi Road, Sucheng District, Suqian, 223800, Jiangsu, People's Republic of China
| | - Qingqing Ye
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, No. 120, Suzhi Road, Sucheng District, Suqian, 223800, Jiangsu, People's Republic of China
| | - Xiaohu Su
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, No. 120, Suzhi Road, Sucheng District, Suqian, 223800, Jiangsu, People's Republic of China
| | - Shuai Shen
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, No. 120, Suzhi Road, Sucheng District, Suqian, 223800, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Wong ET, Lok E. Body Fluids Modulate Propagation of Tumor Treating Fields. Adv Radiat Oncol 2024; 9:101316. [PMID: 38260214 PMCID: PMC10801649 DOI: 10.1016/j.adro.2023.101316] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/13/2023] [Indexed: 01/24/2024] Open
Abstract
Tumor treating fields (TTFields) are nonionizing alternating electric fields that have anticancer properties. After the initial approval for use in patients with recurrent glioblastoma in 2011 and newly diagnosed glioblastomas in 2015, they are now being tested in those with advanced lung cancer, ovarian carcinoma, and pancreatic cancer. Unlike ionizing radiation therapy, TTFields have nonlinear propagation characteristics; therefore, it is difficult for clinicians to recognize intuitively the location where these fields have the most impact. However, finite element analysis offers a means of delineating TTFields in the human body. Our analyses in the brain, pelvis, and thorax revealed that cerebrospinal fluid, edema, urine, ascites, pleural fluid, and necrotic core within a tumor greatly influence their distribution within these body cavities. Our observations thus provided a unified framework on the role of these compartmentalized fluids in influencing the propagation of TTFields.
Collapse
Affiliation(s)
- Eric T. Wong
- Division of Hematology/Oncology, Rhode Island Hospital & Lifespan Cancer Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Edwin Lok
- Division of Hematology/Oncology, Rhode Island Hospital & Lifespan Cancer Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
28
|
Slavkov D, Hadzhiyanev A, Slavkova S. Tumor treating fields: a new treatment for glioblastoma. BIOTECHNOL BIOTEC EQ 2023. [DOI: 10.1080/13102818.2022.2155567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Dimitar Slavkov
- Clinic for Neurosurgery, Spine Surgery and Neuromodulation, HELIOS Vogtland-Klinikum Plauen GmbH, Plauen, Germany
| | - Asen Hadzhiyanev
- Department of Neurosurgery, University Hospital ‘St. Ivan Rilsky’, Medical University of Sofia, Sofia, Bulgaria
| | - Svetoslava Slavkova
- Clinic for Dermatology, HELIOS Vogtland-Klinikum Plauen GmbH, Plauen, Germany
| |
Collapse
|
29
|
Xiang XW, Liu HT, Tao XN, Zeng YL, Liu J, Wang C, Yu SX, Zhao H, Liu YJ, Liu KF. Glioblastoma behavior study under different frequency electromagnetic field. iScience 2023; 26:108575. [PMID: 38125027 PMCID: PMC10730381 DOI: 10.1016/j.isci.2023.108575] [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: 06/04/2023] [Revised: 10/06/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
The tumor-treating fields (TTFields) technology has revolutionized the management of recurrent and newly diagnosed glioblastoma (GBM) cases. To ameliorate this treatment modality for GBM and other oncological conditions, it is necessary to understand the biophysical principles of TTFields better. In this study, we further analyzed the mechanism of the electromagnetic exposure with varying frequencies and electric field strengths on cells in mitosis, specifically in telophase. In reference to previous studies, an intuitive finite element model of the mitotic cell was built for electromagnetic simulations, predicting a local increase in the cleavage furrow region, which may help explain TTFields' anti-proliferative effects. Cell experiments confirmed that the reduction in proliferation and migration of glioma cell by TTFields was in a frequency- and field-strength-dependent manner. This work provides unique insights into the selection of frequencies in the anti-proliferative effect of TTFields on tumors, which could improve the application of TTFields.
Collapse
Affiliation(s)
- Xiao-Wei Xiang
- Academy for engineering & technology, Fudan University, Shanghai 200433, China
| | - Hao-Tian Liu
- Academy for engineering & technology, Fudan University, Shanghai 200433, China
| | - Xiao-Nan Tao
- School of information science and technology, Fudan University, Shanghai 200433, China
| | - Yu-Lian Zeng
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Jing Liu
- School of information science and technology, Fudan University, Shanghai 200433, China
| | - Chen Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hui Zhao
- School of information science and technology, Fudan University, Shanghai 200433, China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ke-Fu Liu
- School of information science and technology, Fudan University, Shanghai 200433, China
| |
Collapse
|
30
|
Olatunji G, Aderinto N, Adefusi T, Kokori E, Akinmoju O, Yusuf I, Olusakin T, Muzammil MA. Efficacy of tumour-treating fields therapy in recurrent glioblastoma: A narrative review of current evidence. Medicine (Baltimore) 2023; 102:e36421. [PMID: 38050252 PMCID: PMC10695547 DOI: 10.1097/md.0000000000036421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023] Open
Abstract
Recurrent Glioblastoma presents a formidable challenge in oncology due to its aggressive nature and limited treatment options. Tumour-Treating Fields (TTFields) Therapy, a novel therapeutic modality, has emerged as a promising approach to address this clinical conundrum. This review synthesizes the current evidence surrounding the efficacy of TTFields Therapy in the context of recurrent Glioblastoma. Diverse academic databases were explored to identify relevant studies published within the last decade. Strategic keyword selection facilitated the inclusion of studies focusing on TTFields Therapy's efficacy, treatment outcomes, and patient-specific factors. The review reveals a growing body of evidence suggesting the potential clinical benefits of TTFields Therapy for patients with recurrent Glioblastoma. Studies consistently demonstrate its positive impact on overall survival (OS) and progression-free survival (PFS). The therapy's safety profile remains favorable, with mild to moderate skin reactions being the most commonly reported adverse events. Our analysis highlights the importance of patient selection criteria, with emerging biomarkers such as PTEN mutation status influencing therapy response. Additionally, investigations into combining TTFields Therapy with other treatments, including surgical interventions and novel approaches, offer promising avenues for enhancing therapeutic outcomes. The synthesis of diverse studies underscores the potential of TTFields Therapy as a valuable addition to the armamentarium against recurrent Glioblastoma. The narrative review comprehensively explains the therapy's mechanisms, clinical benefits, adverse events, and future directions. The insights gathered herein serve as a foundation for clinicians and researchers striving to optimize treatment strategies for patients facing the challenging landscape of recurrent Glioblastoma.
Collapse
Affiliation(s)
- Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Ismaila Yusuf
- Department of Medicine and Surgery, Obafemi Awolowo University, Ife, Nigeria
| | - Tobi Olusakin
- College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
31
|
Hambarde S, Manalo JM, Baskin DS, Sharpe MA, Helekar SA. Spinning magnetic field patterns that cause oncolysis by oxidative stress in glioma cells. Sci Rep 2023; 13:19264. [PMID: 37935811 PMCID: PMC10630398 DOI: 10.1038/s41598-023-46758-w] [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: 01/13/2023] [Accepted: 11/04/2023] [Indexed: 11/09/2023] Open
Abstract
Raising reactive oxygen species (ROS) levels in cancer cells to cause macromolecular damage and cell death is a promising anticancer treatment strategy. Observations that electromagnetic fields (EMF) elevate intracellular ROS and cause cancer cell death, have led us to develop a new portable wearable EMF device that generates spinning oscillating magnetic fields (sOMF) to selectively kill cancer cells while sparing normal cells in vitro and to shrink GBM tumors in vivo through a novel mechanism. Here, we characterized the precise configurations and timings of sOMF stimulation that produce cytotoxicity due to a critical rise in superoxide in two types of human glioma cells. We also found that the antioxidant Trolox reverses the cytotoxic effect of sOMF on glioma cells indicating that ROS play a causal role in producing the effect. Our findings clarify the link between the physics of magnetic stimulation and its mechanism of anticancer action, facilitating the development of a potential new safe noninvasive device-based treatment for GBM and other gliomas.
Collapse
Affiliation(s)
- Shashank Hambarde
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
| | - Jeanne M Manalo
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
| | - David S Baskin
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
- Department of Neurosurgery, Weill Cornell Medical College, New York, NY, USA
| | - Martyn A Sharpe
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
| | - Santosh A Helekar
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Houston Methodist Hospital, Houston, TX, USA.
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA.
- Houston Methodist Research Institute, Houston, TX, USA.
- Department of Neurosurgery, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
32
|
Kumaria A, Ashkan K. Novel therapeutic strategies in glioma targeting glutamatergic neurotransmission. Brain Res 2023; 1818:148515. [PMID: 37543066 DOI: 10.1016/j.brainres.2023.148515] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/11/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023]
Abstract
High grade gliomas carry a poor prognosis despite aggressive surgical and adjuvant approaches including chemoradiotherapy. Recent studies have demonstrated a mitogenic association between neuronal electrical activity and glioma growth involving the PI3K-mTOR pathway. As the predominant excitatory neurotransmitter of the brain, glutamate signalling in particular has been shown to promote glioma invasion and growth. The concept of the neurogliomal synapse has been established whereby glutamatergic receptors on glioma cells have been shown to promote tumour propagation. Targeting glutamatergic signalling is therefore a potential treatment option in glioma. Antiepileptic medications decrease excess neuronal electrical activity and some may possess anti-glutamate effects. Although antiepileptic medications continue to be investigated for an anti-glioma effect, good quality randomised trial evidence is lacking. Other pharmacological strategies that downregulate glutamatergic signalling include riluzole, memantine and anaesthetic agents. Neuromodulatory interventions possessing potential anti-glutamate activity include deep brain stimulation and vagus nerve stimulation - this contributes to the anti-seizure efficacy of the latter and the possible neuroprotective effect of the former. A possible role of neuromodulation as a novel anti-glioma modality has previously been proposed and that hypothesis is extended to include these modalities. Similarly, the significant survival benefit in glioblastoma attributable to alternating electrical fields (Tumour Treating Fields) may be a result of disruption to neurogliomal signalling. Further studies exploring excitatory neurotransmission and glutamatergic signalling and their role in glioma origin, growth and propagation are therefore warranted.
Collapse
Affiliation(s)
- Ashwin Kumaria
- Department of Neurosurgery, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK.
| | | |
Collapse
|
33
|
Mikobi EK, Voorhees G, Mahmood A, Gandhi A, Bailey M, Phillips J. Influence of Tumor-Treating Fields in a Young Patient With Primary Spinal Glioblastoma Multiforme (GBM): A Case Report of a Rare Tumor. Cureus 2023; 15:e49441. [PMID: 38149153 PMCID: PMC10750880 DOI: 10.7759/cureus.49441] [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] [Accepted: 11/17/2023] [Indexed: 12/28/2023] Open
Abstract
We present a 31-year-old female patient with primary glioblastoma multiforme (GBM) of the thoracic spine, diagnosed in approximately mid-2020. Her symptoms began several months prior with right foot paresthesia, which progressed to neuropathy ascending from her distal to proximal right lower extremity. Over several months, she developed lumbo-thoracic throbbing pain, which was dermatomal radiating anteriorly. Her pain worsened with activity. A thoracic spine MRI showed a focus of abnormal intradural intramedullary enhancement present from the T10-T11 disc level to the T12-L1 disc level, producing a large amount of edema within the cord. She underwent a gross total surgical resection. The patient had WHO Grade IV spinal GBM per histopathology. The patient received adjuvant concurrent radiation therapy and temozolomide chemotherapy. She continues with maintenance temozolomide along with the compassionate use of Novocure alternating electrical field therapy for the spine. She is being monitored closely by a multi-specialty team. At 32 months post-radiation therapy, her disease is stable with no evidence of progression. She has made significant improvements in her ambulation and symptoms. While GBM is most commonly intracranial, primary spinal GBM is relatively rare. Although established treatment guidelines exist for supratentorial GBM, treatment protocol choices for spinal GBM remain controversial but typically mirror those used for intracranial GBM and include surgery, radiation therapy, and chemotherapy. Alternating electrical field therapy, also known as tumor-treating fields (TTFields), is indicated for adjuvant treatment of intracranial GBM. While further studies of TTFields in spinal GBM are needed, TTFields appear to be a safe adjunct treatment for spinal GBM. Further studies are still needed aimed at finding an improved treatment for spinal GBM.
Collapse
Affiliation(s)
- Emmanuel K Mikobi
- Internal Medicine, Corpus Christi Medical Center, Corpus Christi, USA
| | - Gerard Voorhees
- Radiation Oncology, Corpus Christi Medical Center, Corpus Christi, USA
| | - Aftab Mahmood
- Hematology and Oncology, Corpus Christi Medical Center, Corpus Christi, USA
| | - Arpit Gandhi
- Radiology, Radiology Associates, Corpus Christi, USA
| | - Michael Bailey
- Pathology, Corpus Christi Medical Center, Corpus Christi, USA
| | - Jacqueline Phillips
- Internal Medicine, Graduate Medical Education, Corpus Christi Medical Center, Corpus Christi, USA
| |
Collapse
|
34
|
Homami E, Goliaei B, Shariatpanahi SP, Habibi-Kelishomi Z. Alternating electric fields can improve chemotherapy treatment efficacy in blood cancer cell U937 (non-adherent cells). BMC Cancer 2023; 23:861. [PMID: 37700230 PMCID: PMC10496298 DOI: 10.1186/s12885-023-11339-7] [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: 06/14/2023] [Accepted: 08/25/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Recent achievements in cancer therapy are the use of alternating electrical fields at intermediate frequencies (100-300 kHz) and low intensities (1-3 V/cm), which specifically target cell proliferation while affecting different cellular activities depending on the frequency used. METHODS In this article, we examine the effect of electric fields on spherical suspended cells and propose the combination of Daunorubicin, a chemotherapy agent widely used in the treatment of acute myeloid leukemia, with electric field exposure. U937 cells were subjected to an electric field with a frequency of 200 kHz and an intensity of 0.75 V/cm, or to a combination of Daunorubicin and electric field exposure, resulting in a significant reduction in cell proliferation. Furthermore, the application of an electric field to U937 cells increased Daunorubicin uptake. RESULTS Apoptosis and DNA damage were induced by the electric field or in conjunction with Daunorubicin. Notably, normal cells exposed to an electric field did not show significant damage, indicating a selective effect on dividing cancer cells (U937). Moreover, the electric field affects the U937 cell line either alone or in combination with Daunorubicin. This effect may be due to increased membrane permeability. CONCLUSIONS Our findings suggest that the use of electric fields at intermediate frequencies and low intensities, either alone or in combination with Daunorubicin, has potential as a selective anti-cancer therapy for dividing cancer cells, particularly in the treatment of acute myeloid leukemia. Further research is needed to fully understand the underlying mechanisms and to optimize the use of this therapy.
Collapse
Affiliation(s)
- Elham Homami
- Institute of Biochemistry and Biophysics, University of Tehran, PO Box 13145-1384, Tehran, Iran
| | - Bahram Goliaei
- Institute of Biochemistry and Biophysics, University of Tehran, PO Box 13145-1384, Tehran, Iran.
| | | | - Zahra Habibi-Kelishomi
- Institute of Biochemistry and Biophysics, University of Tehran, PO Box 13145-1384, Tehran, Iran
| |
Collapse
|
35
|
Li X, Liu K, Xing L, Rubinsky B. A review of tumor treating fields (TTFields): advancements in clinical applications and mechanistic insights. Radiol Oncol 2023; 57:279-291. [PMID: 37665740 PMCID: PMC10476910 DOI: 10.2478/raon-2023-0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/04/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Tumor Treating Fields (TTFields) is a non-invasive modality for cancer treatment that utilizes a specific sinusoidal electric field ranging from 100 kHz to 300 kHz, with an intensity of 1 V/cm to 3 V/cm. Its purpose is to inhibit cancer cell proliferation and induce cell death. Despite promising outcomes from clinical trials, TTFields have received FDA approval for the treatment of glioblastoma multiforme (GBM) and malignant pleural mesothelioma (MPM). Nevertheless, global acceptance of TTFields remains limited. To enhance its clinical application in other types of cancer and gain a better understanding of its mechanisms of action, this review aims to summarize the current research status by examining existing literature on TTFields' clinical trials and mechanism studies. CONCLUSIONS Through this comprehensive review, we seek to stimulate novel ideas and provide physicians, patients, and researchers with a better comprehension of the development of TTFields and its potential applications in cancer treatment.
Collapse
Affiliation(s)
- Xing Li
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nan Jing, Jiang Su, China
| | - Kaida Liu
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nan Jing, Jiang Su, China
| | - Lidong Xing
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nan Jing, Jiang Su, China
| | - Boris Rubinsky
- Department of Mechanical Engineering, University of California Berkeley, BerkeleyCA, United States of America
| |
Collapse
|
36
|
Guterres A, Abrahim M, da Costa Neves PC. The role of immune subtyping in glioma mRNA vaccine development. Immunotherapy 2023; 15:1057-1072. [PMID: 37431617 DOI: 10.2217/imt-2023-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023] Open
Abstract
Studies on the development of mRNA vaccines for central nervous system tumors have used gene expression profiles, clinical data and RNA sequencing from sources such as The Cancer Genome Atlas and Chinese Glioma Genome Atlas to identify effective antigens. These studies revealed several immune subtypes of glioma, each one linked to unique prognoses and genetic/immune-modulatory changes. Potential antigens include ARPC1B, BRCA2, COL6A1, ITGB3, IDH1, LILRB2, TP53 and KDR, among others. Patients with immune-active and immune-suppressive phenotypes were found to respond better to mRNA vaccines. While these findings indicate the potential of mRNA vaccines in cancer therapy, further research is required to optimize administration and adjuvant selection, and precisely identify target antigens.
Collapse
Affiliation(s)
- Alexandro Guterres
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, 21040-360, Brazil
| | - Mayla Abrahim
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, 21040-360, Brazil
| | - Patrícia Cristina da Costa Neves
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, 21040-360, Brazil
| |
Collapse
|
37
|
Jones AB, Schanel TL, Rigsby MR, Griguer CE, McFarland BC, Anderson JC, Willey CD, Hjelmeland AB. Tumor Treating Fields Alter the Kinomic Landscape in Glioblastoma Revealing Therapeutic Vulnerabilities. Cells 2023; 12:2171. [PMID: 37681903 PMCID: PMC10486683 DOI: 10.3390/cells12172171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Treatment for the deadly brain tumor glioblastoma (GBM) has been improved through the non-invasive addition of alternating electric fields, called tumor treating fields (TTFields). Improving both progression-free and overall survival, TTFields are currently approved for treatment of recurrent GBMs as a monotherapy and in the adjuvant setting alongside TMZ for newly diagnosed GBMs. These TTFields are known to inhibit mitosis, but the full molecular impact of TTFields remains undetermined. Therefore, we sought to understand the ability of TTFields to disrupt the growth patterns of and induce kinomic landscape shifts in TMZ-sensitive and -resistant GBM cells. We determined that TTFields significantly decreased the growth of TMZ-sensitive and -resistant cells. Kinomic profiling predicted kinases that were induced or repressed by TTFields, suggesting possible therapy-specific vulnerabilities. Serving as a potential pro-survival mechanism for TTFields, kinomics predicted the increased activity of platelet-derived growth-factor receptor alpha (PDGFRα). We demonstrated that the addition of the PDGFR inhibitor, crenolanib, to TTFields further reduced cell growth in comparison to either treatment alone. Collectively, our data suggest the efficacy of TTFields in vitro and identify common signaling responses to TTFields in TMZ-sensitive and -resistant populations, which may support more personalized medicine approaches.
Collapse
Affiliation(s)
- Amber B. Jones
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.B.J.); (M.R.R.); (B.C.M.)
| | - Taylor L. Schanel
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (T.L.S.); (J.C.A.)
| | - Mikayla R. Rigsby
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.B.J.); (M.R.R.); (B.C.M.)
| | - Corinne E. Griguer
- Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA;
| | - Braden C. McFarland
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.B.J.); (M.R.R.); (B.C.M.)
| | - Joshua C. Anderson
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (T.L.S.); (J.C.A.)
| | - Christopher D. Willey
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (T.L.S.); (J.C.A.)
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.B.J.); (M.R.R.); (B.C.M.)
| |
Collapse
|
38
|
Lee WS, Jang Y, Cho A, Kim YB, Bu YH, Yang S, Kim EH. Effectiveness of tumor‑treating fields to reduce the proliferation and migration of liposarcoma cell lines. Exp Ther Med 2023; 26:363. [PMID: 37408858 PMCID: PMC10318604 DOI: 10.3892/etm.2023.12062] [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: 10/26/2022] [Accepted: 02/21/2023] [Indexed: 07/07/2023] Open
Abstract
Liposarcoma (LPS) is a rare type of soft tissue sarcoma that constitutes 20% of all sarcoma cases in adults. Effective therapeutic protocols for human LPS are not well-defined. Tumor-treating fields (TTFields) are a novel and upcoming field for antitumor therapy. TTFields combined with chemoradiotherapy have proven to be more effective than TTFields combined with radiotherapy or chemotherapy alone. The present study aimed to assess the effectiveness of TTFields in inhibiting cell proliferation and viability for the anticancer treatment of LPS. The present study used TTFields (frequency, 150 kHz; intensity, 1.0 V/cm) to treat two LPS cell lines (94T778 and SW872) and analyzed the antitumor effects. According to trypan blue and MTT assay results, TTFields markedly reduced the viability and proliferation of LPS cell lines along with the formation of colonies in three-dimensional culture. Based on the Transwell chamber assay, TTFields treatment also markedly reduced the migration of LPS cells. Furthermore, as shown by the higher activation of caspase-3 in the Caspase-3 activity assay and the results of the reactive oxygen species (ROS) assay, TTFields increased the formation of ROS in the cells and enhanced the proportion of apoptotic cells. The present study also investigated the inhibitory effect of TTFields in combination with doxorubicin (DOX) on the migratory capacity of tumor cells. The results demonstrated that TTFields treatment synergistically induced the ROS-induced apoptosis of LPS cancer cell lines and inhibited their migratory behavior. In conclusion, the present study demonstrated the potential of TTFields in improving the sensitivity of LPS cancer cells, which may lay the foundation for future clinical trials of this combination treatment strategy.
Collapse
Affiliation(s)
- Won Seok Lee
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Yoonjung Jang
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Gyeongsangbuk-do 42988, Republic of Korea
| | - Ahyeon Cho
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Yu Bin Kim
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Young Hyun Bu
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Somi Yang
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| |
Collapse
|
39
|
Campana LG, Daud A, Lancellotti F, Arroyo JP, Davalos RV, Di Prata C, Gehl J. Pulsed Electric Fields in Oncology: A Snapshot of Current Clinical Practices and Research Directions from the 4th World Congress of Electroporation. Cancers (Basel) 2023; 15:3340. [PMID: 37444450 PMCID: PMC10340685 DOI: 10.3390/cancers15133340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/29/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The 4th World Congress of Electroporation (Copenhagen, 9-13 October 2022) provided a unique opportunity to convene leading experts in pulsed electric fields (PEF). PEF-based therapies harness electric fields to produce therapeutically useful effects on cancers and represent a valuable option for a variety of patients. As such, irreversible electroporation (IRE), gene electrotransfer (GET), electrochemotherapy (ECT), calcium electroporation (Ca-EP), and tumour-treating fields (TTF) are on the rise. Still, their full therapeutic potential remains underappreciated, and the field faces fragmentation, as shown by parallel maturation and differences in the stages of development and regulatory approval worldwide. This narrative review provides a glimpse of PEF-based techniques, including key mechanisms, clinical indications, and advances in therapy; finally, it offers insights into current research directions. By highlighting a common ground, the authors aim to break silos, strengthen cross-functional collaboration, and pave the way to novel possibilities for intervention. Intriguingly, beyond their peculiar mechanism of action, PEF-based therapies share technical interconnections and multifaceted biological effects (e.g., vascular, immunological) worth exploiting in combinatorial strategies.
Collapse
Affiliation(s)
- Luca G. Campana
- Department of Surgery, Manchester University NHS Foundation Trust, Oxford Rd., Manchester M13 9WL, UK;
| | - Adil Daud
- Department of Medicine, University of California, 550 16 Street, San Francisco, CA 94158, USA;
| | - Francesco Lancellotti
- Department of Surgery, Manchester University NHS Foundation Trust, Oxford Rd., Manchester M13 9WL, UK;
| | - Julio P. Arroyo
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; (J.P.A.); (R.V.D.)
| | - Rafael V. Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA; (J.P.A.); (R.V.D.)
- Institute for Critical Technology and Applied Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Claudia Di Prata
- Department of Surgery, San Martino Hospital, 32100 Belluno, Italy;
| | - Julie Gehl
- Department of Clinical Oncology and Palliative Care, Zealand University Hospital, 4000 Roskilde, Denmark;
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| |
Collapse
|
40
|
Smothers AR, Henderson JR, O'Connell JJ, Stenbeck JM, Dean D, Booth BW. Optimization of tumor-treating field therapy for triple-negative breast cancer cells in vitro via frequency modulation. Cancer Cell Int 2023; 23:110. [PMID: 37287008 DOI: 10.1186/s12935-023-02959-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: 04/17/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023] Open
Abstract
PURPOSE Currently, tumor-treating field (TTField) therapy utilizes a single "optimal" frequency of electric fields to achieve maximal cell death in a targeted population of cells. However, because of differences in cell size, shape, and ploidy during mitosis, optimal electric field characteristics for universal maximal cell death may not exist. This study investigated the anti-mitotic effects of modulating electric field frequency as opposed to utilizing uniform electric fields. METHODS We developed and validated a custom device that delivers a wide variety of electric field and treatment parameters including frequency modulation. We investigated the efficacy of frequency modulating tumor-treating fields on triple-negative breast cancer cells compared to human breast epithelial cells. RESULTS We show that frequency-modulated (FM) TTFields are as selective at treating triple-negative breast cancer (TNBC) as uniform TTFields while having a greater efficacy for combating TNBC cell growth. TTField treatment at a mean frequency of 150 kHz with a frequency range of ± 10 kHz induced apoptosis in a greater number of TNBC cells after 24 h as compared to unmodulated treatment which led to further decreased cell viability after 48 h. Furthermore, all TNBC cells died after 72 h of FM treatment while cells that received unmodulated treatment were able to recover to cell number equivalent to the control. CONCLUSION TTFields were highly efficacious against TNBC growth, FM TTFields showed minimal effects on epithelial cells similar to unmodulated treatment.
Collapse
Affiliation(s)
- Austin R Smothers
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson University, Clemson, SC, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | | | - John J O'Connell
- Prisma Health Cancer Institute, Prisma Health, Greenville, SC, USA
- Clemson University School of Health Research, Clemson, SC, USA
- University of South Carolina School of Medicine-Greenville, Greenville, SC, USA
| | | | - Delphine Dean
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson University, Clemson, SC, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Brian W Booth
- Department of Bioengineering, Clemson University, Clemson, SC, USA.
| |
Collapse
|
41
|
Regnery S, Franke H, Held T, Trinh T, Naveh A, Abraham Y, Hörner-Rieber J, Hess J, Huber PE, Debus J, Lopez Perez R, Adeberg S. Tumor treating fields as novel combination partner in the multimodal treatment of head and neck cancer. Head Neck 2023; 45:838-848. [PMID: 36872620 DOI: 10.1002/hed.27298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/20/2022] [Accepted: 01/23/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND We aimed to demonstrate the effects of tumor treating fields (TTFields) in head and neck squamous cell carcinoma (HNSCC) cells when combined with radiotherapy (RT) and chemotherapy. METHODS Two human HNSCC cell lines (Cal27, FaDu) received five different treatments: TTFields, RT +/- TTFields and RT + simultaneous cisplatin +/- TTFields. Effects were quantified using clonogenic assays and flow cytometric analyses of DAPI, caspase-3 activation and γH2AX foci. RESULTS Treatment with RT + TTFields decreased the clonogenic survival as strong as treatment with RT + simultaneous cisplatin. The triple combination of RT + simultaneous cisplatin + TTFields even further decreased the clonogenic survival. Accordingly, combination of TTFields with RT or RT + simultaneous cisplatin increased cellular apoptosis and DNA double-strand breaks. CONCLUSION TTFields therapy seems a promising combination partner in the multimodal treatment of locally advanced HNSCC. It could be used to intensify chemoradiotherapy or as alternative to chemotherapy.
Collapse
Affiliation(s)
- Sebastian Regnery
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Henrik Franke
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Held
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thuy Trinh
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Juliane Hörner-Rieber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jochen Hess
- Section of Experimental and Translation Head and neck Oncology, Department of Otorhinolaryngology, Heidelberg University Hospital, Heidelberg, Germany.,Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ramon Lopez Perez
- Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Adeberg
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
42
|
Wang X, Gong Z, Wang T, Law J, Chen X, Wanggou S, Wang J, Ying B, Francisco M, Dong W, Xiong Y, Fan JJ, MacLeod G, Angers S, Li X, Dirks PB, Liu X, Huang X, Sun Y. Mechanical nanosurgery of chemoresistant glioblastoma using magnetically controlled carbon nanotubes. SCIENCE ADVANCES 2023; 9:eade5321. [PMID: 36989359 PMCID: PMC10058241 DOI: 10.1126/sciadv.ade5321] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain cancer. Despite multimodal treatment including surgery, radiotherapy, and chemotherapy, median patient survival has remained at ~15 months for decades. This situation demands an outside-the-box treatment approach. Using magnetic carbon nanotubes (mCNTs) and precision magnetic field control, we report a mechanical approach to treat chemoresistant GBM. We show that GBM cells internalize mCNTs, the mobilization of which by rotating magnetic field results in cell death. Spatiotemporally controlled mobilization of intratumorally delivered mCNTs suppresses GBM growth in vivo. Functionalization of mCNTs with anti-CD44 antibody, which recognizes GBM cell surface-enriched antigen CD44, increases mCNT recognition of cancer cells, prolongs mCNT enrichment within the tumor, and enhances therapeutic efficacy. Using mouse models of GBM with upfront or therapy-induced resistance to temozolomide, we show that mCNT treatment is effective in treating chemoresistant GBM. Together, we establish mCNT-based mechanical nanosurgery as a treatment option for GBM.
Collapse
Affiliation(s)
- Xian Wang
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zheyuan Gong
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Tiancong Wang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Xin Chen
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Siyi Wanggou
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jintian Wang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Binbin Ying
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Michelle Francisco
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Weifan Dong
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Yi Xiong
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jerry J. Fan
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Graham MacLeod
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Stephane Angers
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peter B. Dirks
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Xinyu Liu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Xi Huang
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Corresponding author. (X.H.); (Y.S.)
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Corresponding author. (X.H.); (Y.S.)
| |
Collapse
|
43
|
Zhou Y, Xing X, Zhou J, Jiang H, Cen P, Jin C, Zhong Y, Zhou R, Wang J, Tian M, Zhang H. Therapeutic potential of tumor treating fields for malignant brain tumors. Cancer Rep (Hoboken) 2023; 6:e1813. [PMID: 36987739 PMCID: PMC10172187 DOI: 10.1002/cnr2.1813] [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: 01/18/2023] [Revised: 03/02/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Malignant brain tumors are among the most threatening diseases of the central nervous system, and despite increasingly updated treatments, the prognosis has not been improved. Tumor treating fields (TTFields) are an emerging approach in cancer treatment using intermediate-frequency and low-intensity electric field and can lead to the development of novel therapeutic options. RECENT FINDINGS A series of biological processes induced by TTFields to exert anti-cancer effects have been identified. Recent studies have shown that TTFields can alter the bioelectrical state of macromolecules and organelles involved in cancer biology. Massive alterations in cancer cell proteomics and transcriptomics caused by TTFields were related to cell biological processes as well as multiple organelle structures and activities. This review addresses the mechanisms of TTFields and recent advances in the application of TTFields therapy in malignant brain tumors, especially in glioblastoma (GBM). CONCLUSIONS As a novel therapeutic strategy, TTFields have shown promising results in many clinical trials, especially in GBM, and continue to evolve. A growing number of patients with malignant brain tumors are being enrolled in ongoing clinical studies demonstrating that TTFields-based combination therapies can improve treatment outcomes.
Collapse
Affiliation(s)
- Youyou Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiaoqing Xing
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinyun Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Han Jiang
- Faculty of Science and Technology, Department of Electrical and Computer Engineering, Biomedical Imaging Laboratory (BIG), University of Macau, Taipa, Macau SAR, China
| | - Peili Cen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
44
|
Smothers AR, Henderson JR, O'Connell JJ, Stenbeck JM, Dean D, Harvey TG, Booth BW. Efficacy and selectivity of tumor-treating field therapy for triple-negative breast cancer cells via in-house delivery device. Discov Oncol 2023; 14:34. [PMID: 36991198 PMCID: PMC10060613 DOI: 10.1007/s12672-023-00647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
PURPOSE Triple-negative breast cancer continues to be one of the leading causes of death in women, making up 7% of all cancer deaths. Tumor-treating electric fields are low-energy, low-frequency oscillating electric fields that induce an anti-proliferative effect on mitotic cells in glioblastoma multiforme, non-small cell lung cancer, and ovarian cancer. Little is known about effects of tumor-treating fields on triple-negative breast cancer and known research for tumor-treating fields only utilizes low (< 3 V/cm) electric field intensities. METHODS We have developed an in-house field delivery device capable of high levels of customization to explore a much wider variety of electric field and treatment parameters. Furthermore, we investigated the selectivity of tumor-treating field treatment between triple-negative breast cancer and human breast epithelial cells. RESULTS Tumor-treating fields show greatest efficacy against triple-negative breast cancer cell lines between 1 and 3 V/cm electric field intensities while having little effect on epithelial cells. CONCLUSION These results provide a clear therapeutic window for tumor-treating field delivery to triple-negative breast cancer.
Collapse
Affiliation(s)
- Austin R Smothers
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson University, Clemson, SC, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | | | - John J O'Connell
- Prisma Health Cancer Institute, Prisma Health, Greenville, SC, USA
- Clemson University School of Health Research, Clemson, SC, USA
- University of South Carolina School of Medicine-Greenville, Greenville, SC, USA
| | | | - Delphine Dean
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson University, Clemson, SC, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Tyler G Harvey
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Brian W Booth
- Department of Bioengineering, Clemson University, Clemson, SC, USA.
| |
Collapse
|
45
|
Abed T, Ganser K, Eckert F, Stransky N, Huber SM. Ion channels as molecular targets of glioblastoma electrotherapy. Front Cell Neurosci 2023; 17:1133984. [PMID: 37006466 PMCID: PMC10064067 DOI: 10.3389/fncel.2023.1133984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/10/2023] [Indexed: 03/19/2023] Open
Abstract
Therapies with weak, non-ionizing electromagnetic fields comprise FDA-approved treatments such as Tumor Treating Fields (TTFields) that are used for adjuvant therapy of glioblastoma. In vitro data and animal models suggest a variety of biological TTFields effects. In particular, effects ranging from direct tumoricidal, radio- or chemotherapy-sensitizing, metastatic spread-inhibiting, up to immunostimulation have been described. Diverse underlying molecular mechanisms, such as dielectrophoresis of cellular compounds during cytokinesis, disturbing the formation of the spindle apparatus during mitosis, and perforating the plasma membrane have been proposed. Little attention, however, has been paid to molecular structures that are predestinated to percept electromagnetic fields-the voltage sensors of voltage-gated ion channels. The present review article briefly summarizes the mode of action of voltage sensing by ion channels. Moreover, it introduces into the perception of ultra-weak electric fields by specific organs of fishes with voltage-gated ion channels as key functional units therein. Finally, this article provides an overview of the published data on modulation of ion channel function by diverse external electromagnetic field protocols. Combined, these data strongly point to a function of voltage-gated ion channels as transducers between electricity and biology and, hence, to voltage-gated ion channels as primary targets of electrotherapy.
Collapse
Affiliation(s)
- Tayeb Abed
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Radiation Oncology, Medical University Vienna, Vienna, Austria
| | - Nicolai Stransky
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
46
|
Tumor Treating Fields (TTFields) Therapy Concomitant with Taxanes for Cancer Treatment. Cancers (Basel) 2023; 15:cancers15030636. [PMID: 36765594 PMCID: PMC9913762 DOI: 10.3390/cancers15030636] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Non-small cell lung cancer, ovarian cancer, and pancreatic cancer all present with high morbidity and mortality. Systemic chemotherapies have historically been the cornerstone of standard of care (SOC) regimens for many cancers, but are associated with systemic toxicity. Multimodal treatment combinations can help improve patient outcomes; however, implementation is limited by additive toxicities and potential drug-drug interactions. As such, there is a high unmet need to develop additional therapies to enhance the efficacy of SOC treatments without increasing toxicity. Tumor Treating Fields (TTFields) are electric fields that exert physical forces to disrupt cellular processes critical for cancer cell viability and tumor progression. The therapy is locoregional and is delivered noninvasively to the tumor site via a portable medical device that consists of field generator and arrays that are placed on the patient's skin. As a noninvasive treatment modality, TTFields therapy-related adverse events mainly consist of localized skin reactions, which are manageable with effective acute and prophylactic treatments. TTFields selectively target cancer cells through a multi-mechanistic approach without affecting healthy cells and tissues. Therefore, the application of TTFields therapy concomitant with other cancer treatments may lead to enhanced efficacy, with low risk of further systemic toxicity. In this review, we explore TTFields therapy concomitant with taxanes in both preclinical and clinical settings. The summarized data suggest that TTFields therapy concomitant with taxanes may be beneficial in the treatment of certain cancers.
Collapse
|
47
|
Shams S, Patel CB. Anti-cancer mechanisms of action of therapeutic alternating electric fields (tumor treating fields [TTFields]). J Mol Cell Biol 2022; 14:mjac047. [PMID: 35973687 PMCID: PMC9912101 DOI: 10.1093/jmcb/mjac047] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/11/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Despite improved survival outcomes across many cancer types, the prognosis remains grim for certain solid organ cancers including glioblastoma and pancreatic cancer. Invariably in these cancers, the control achieved by time-limited interventions such as traditional surgical resection, radiation therapy, and chemotherapy is short-lived. A new form of anti-cancer therapy called therapeutic alternating electric fields (AEFs) or tumor treating fields (TTFields) has been shown, either by itself or in combination with chemotherapy, to have anti-cancer effects that translate to improved survival outcomes in patients. Although the pre-clinical and clinical data are promising, the mechanisms of TTFields are not fully elucidated. Many investigations are underway to better understand how and why TTFields is able to selectively kill cancer cells and impede their proliferation. The purpose of this review is to summarize and discuss the reported mechanisms of action of TTFields from pre-clinical studies (both in vitro and in vivo). An improved understanding of how TTFields works will guide strategies focused on the timing and combination of TTFields with other therapies, to further improve survival outcomes in patients with solid organ cancers.
Collapse
Affiliation(s)
- Shadi Shams
- Rowan University School of Osteopathic Medicine, Stratford, NJ 08028, USA
| | - Chirag B Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
48
|
Di Gregorio E, Israel S, Staelens M, Tankel G, Shankar K, Tuszyński JA. The distinguishing electrical properties of cancer cells. Phys Life Rev 2022; 43:139-188. [PMID: 36265200 DOI: 10.1016/j.plrev.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
In recent decades, medical research has been primarily focused on the inherited aspect of cancers, despite the reality that only 5-10% of tumours discovered are derived from genetic causes. Cancer is a broad term, and therefore it is inaccurate to address it as a purely genetic disease. Understanding cancer cells' behaviour is the first step in countering them. Behind the scenes, there is a complicated network of environmental factors, DNA errors, metabolic shifts, and electrostatic alterations that build over time and lead to the illness's development. This latter aspect has been analyzed in previous studies, but how the different electrical changes integrate and affect each other is rarely examined. Every cell in the human body possesses electrical properties that are essential for proper behaviour both within and outside of the cell itself. It is not yet clear whether these changes correlate with cell mutation in cancer cells, or only with their subsequent development. Either way, these aspects merit further investigation, especially with regards to their causes and consequences. Trying to block changes at various levels of occurrence or assisting in their prevention could be the key to stopping cells from becoming cancerous. Therefore, a comprehensive understanding of the current knowledge regarding the electrical landscape of cells is much needed. We review four essential electrical characteristics of cells, providing a deep understanding of the electrostatic changes in cancer cells compared to their normal counterparts. In particular, we provide an overview of intracellular and extracellular pH modifications, differences in ionic concentrations in the cytoplasm, transmembrane potential variations, and changes within mitochondria. New therapies targeting or exploiting the electrical properties of cells are developed and tested every year, such as pH-dependent carriers and tumour-treating fields. A brief section regarding the state-of-the-art of these therapies can be found at the end of this review. Finally, we highlight how these alterations integrate and potentially yield indications of cells' malignancy or metastatic index.
Collapse
Affiliation(s)
- Elisabetta Di Gregorio
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Simone Israel
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Michael Staelens
- Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada
| | - Gabriella Tankel
- Department of Mathematics & Statistics, McMaster University, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada
| | - Karthik Shankar
- Department of Electrical & Computer Engineering, University of Alberta, 9211 116 Street NW, Edmonton, T6G 1H9, AB, Canada
| | - Jack A Tuszyński
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada; Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, T6G 1Z2, AB, Canada.
| |
Collapse
|
49
|
Bioelectronic medicines: Therapeutic potential and advancements in next-generation cancer therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188808. [DOI: 10.1016/j.bbcan.2022.188808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/07/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
|
50
|
Moser JC, Salvador E, Deniz K, Swanson K, Tuszynski J, Carlson KW, Karanam NK, Patel CB, Story M, Lou E, Hagemann C. The Mechanisms of Action of Tumor Treating Fields. Cancer Res 2022; 82:3650-3658. [PMID: 35839284 PMCID: PMC9574373 DOI: 10.1158/0008-5472.can-22-0887] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/24/2022] [Accepted: 07/13/2022] [Indexed: 01/07/2023]
Abstract
Tumor treating fields (TTFields), a new modality of cancer treatment, are electric fields transmitted transdermally to tumors. The FDA has approved TTFields for the treatment of glioblastoma multiforme and mesothelioma, and they are currently under study in many other cancer types. While antimitotic effects were the first recognized biological anticancer activity of TTFields, data have shown that tumor treating fields achieve their anticancer effects through multiple mechanisms of action. TTFields therefore have the ability to be useful for many cancer types in combination with many different treatment modalities. Here, we review the current understanding of TTFields and their mechanisms of action.
Collapse
Affiliation(s)
- Justin C. Moser
- HonorHealth Research and Innovation Institute, Scottsdale, Arizona.,Department of Medicine, University of Arizona College of Medicine- Phoenix, Phoenix, Arizona.,Corresponding Author: Justin Moser, HonorHealth Research and Innovation Institute, 10510 N 92nd Street Ste 200, Scottsdale, AZ 85258. Phone: 480-323-4638, E-mail:
| | - Ellaine Salvador
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Karina Deniz
- Department of Medicine, Division of Hematology Oncology and Transplant, University of Minnesota, Minneapolis, Minnesota
| | - Kenneth Swanson
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Jack Tuszynski
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Kristen W. Carlson
- Department of Neurosurgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts
| | - Narasimha Kumar Karanam
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chirag B. Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston Texas.,Neuroscience and Cancer Biology Graduate Programs, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences
| | - Michael Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Emil Lou
- Department of Medicine, Division of Hematology Oncology and Transplant, University of Minnesota, Minneapolis, Minnesota
| | - Carsten Hagemann
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| |
Collapse
|