1
|
Peng J, Zhang L, Wu Y, Chen L, Liu Z, Liu F, Zeng Y. Tumor treating fields may improve the prognosis of patients with lung cancer brain metastases. Brain Stimul 2025; 18:707-709. [PMID: 40112913 DOI: 10.1016/j.brs.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Junsong Peng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Jiangxi. (National Regional Center for Neurological Diseases), Nanchang, Jiangxi, PR China
| | - Yinghua Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Ling Chen
- Department of Neurosurgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
Jobson I, Vo NTN, Kujawinski E, Denning C, Stolnik S, Chauhan VM, Rawson F. Advancing cancer therapy with custom-built alternating electric field devices. Bioelectron Med 2025; 11:2. [PMID: 39881409 PMCID: PMC11780810 DOI: 10.1186/s42234-024-00164-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND In glioblastoma (GBM) therapy research, tumour treating fields by the company Novocure™, have shown promise for increasing patient overall survival. When used with the chemotherapeutic agent temozolomide, they extend median survival by five months. However, there is a space to design alternative systems that will be amenable for wider use in current research. Therefore, we sought to establish a custom-built alternating electric field device to investigate the effect of electrode design on the responsiveness of cancer cells to this therapy. METHODS A 96-well microtiter plate modified with an electrode array was fabricated to investigate its application as an in vitro alternating electric field device. This was initially performed with patient-derived GCE 31 and GIN 31 cell lines found in the core and invasive margin of the GBM tumour, respectively. We sought to establish the effect of the application of low-intensity (3 V/ cm) electric fields with an application duration of 4-48 h, using intermediate frequency (300 kHz) alternating currents (AC). To demonstrate that electric fields were entering the cell, GCE 31 and GIN 31 cells were treated with the inorganic, non-conductive zinc oxide (ZnO) nanoparticles (NP), previously demonstrated to enhance the efficacy of TTFs. After a 4-h exposure to NP, cells were then exposed to alternating electric fields or currents and their metabolic activity was assessed. To better understand how the position and morphology of cells can affect cell therapy responsiveness to alternating electric fields or currents, GBM results were compared to those from the semi-adherent brain tumour cell line, D425. RESULTS Contrary to previous findings, there was no significant difference between the GIN 31 and GCE 31 cells exposed to alternating electric fields or currents treated with or without NP compared to cells untreated and unstimulated. D425 cells exposed to alternating electric fields exhibited a pronounced metabolic increase (1.8-fold), while those exposed to alternating electric currents with or without ZnO had a reduced metabolism relative to the untreated control. CONCLUSIONS The initial hypothesis for the lack of effect of electrical stimulation on the adherent cells was that, due to only a single pair of electrodes being used, the proportion of cells that were in the correct orientation for electric field effects was limited. However, the dramatic shift in cell behaviour of the semi-adherent cells shows that cell morphology plays an important role in the responsiveness of cancer cells to AC electric fields. This study highlights the lack of understanding of the complex mechanisms by which electric fields exert effects on cancer cells. We propose that, for the therapy to be enhanced for patients, research should first focus on the underlying mechanisms of action, specifically on how individual cancer cell types respond to this therapy.
Collapse
Affiliation(s)
- Isobel Jobson
- School of Pharmacy, Biodiscovery Institute & Boots Science Building, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Nguyen T N Vo
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Edward Kujawinski
- Electronic Workshop, Faculty of Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Chris Denning
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Snow Stolnik
- School of Pharmacy, Biodiscovery Institute & Boots Science Building, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Veeren M Chauhan
- School of Pharmacy, Biodiscovery Institute & Boots Science Building, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Frankie Rawson
- School of Pharmacy, Biodiscovery Institute & Boots Science Building, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
7
|
Schlieper-Scherf S, Hebach N, Hausmann D, Azorín DD, Hoffmann DC, Horschitz S, Maier E, Koch P, Karreman MA, Etminan N, Ratliff M. Disrupting glioblastoma networks with tumor treating fields (TTFields) in in vitro models. J Neurooncol 2024; 170:139-151. [PMID: 39088157 PMCID: PMC11457690 DOI: 10.1007/s11060-024-04786-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024]
Abstract
PURPOSE This study investigates the biological effect of Tumor Treating Fields (TTFields) on key drivers of glioblastoma's malignancy-tumor microtube (TM) formation-and on the function and overall integrity of the tumor cell network. METHOD Using a two-dimensional monoculture GB cell network model (2DTM) of primary glioblastoma cell (GBC) cultures (S24, BG5 or T269), we evaluated the effects of TTFields on cell density, interconnectivity and structural integrity of the tumor network. We also analyzed calcium (Ca2+) transient dynamics and network morphology, validating findings in patient-derived tumoroids and brain tumor organoids. RESULTS In the 2DTM assay, TTFields reduced cell density by 85-88% and disrupted network interconnectivity, particularly in cells with multiple TMs. A "crooked TM" phenotype emerged in 5-6% of treated cells, rarely seen in controls. Ca2+ transients were significantly compromised, with global Ca2+ activity reduced by 51-83%, active and periodic cells by over 50%, and intercellular co-activity by 52% in S24, and almost completely in BG5 GBCs. The effects were more pronounced at 200 kHz compared to a 50 kHz TTFields. Similar reductions in Ca2+ activity were observed in patient-derived tumoroids. In brain tumor organoids, TTFields significantly reduced tumor cell proliferation and infiltration. CONCLUSION Our comprehensive study provides new insights into the multiple effects of Inovitro-modeled TTFields on glioma progression, morphology and network dynamics in vitro. Future in vivo studies to verify our in vitro findings may provide the basis for a deeper understanding and optimization of TTFields as a therapeutic modality in the treatment of GB.
Collapse
Affiliation(s)
- Steffen Schlieper-Scherf
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nils Hebach
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - David Hausmann
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel D Azorín
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk C Hoffmann
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Sandra Horschitz
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
- Hector Institute for Translational Brain Research (HITBR gGmbH), Mannheim, Germany
| | - Elena Maier
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Phillip Koch
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
- Hector Institute for Translational Brain Research (HITBR gGmbH), Mannheim, Germany
| | - Matthia A Karreman
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Nima Etminan
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Miriam Ratliff
- Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany.
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
8
|
Strack M, Kückelhaus J, Diebold M, Wuchter P, Huber PE, Schnell O, Sankowski R, Prinz M, Grosu AL, Heiland DH, Nicolay NH, Rühle A. Effects of tumor treating fields (TTFields) on human mesenchymal stromal cells. J Neurooncol 2024; 169:329-340. [PMID: 38900237 PMCID: PMC11341748 DOI: 10.1007/s11060-024-04740-0] [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: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE Mesenchymal stromal cells (MSCs) within the glioblastoma microenvironment have been shown to promote tumor progression. Tumor Treating Fields (TTFields) are alternating electric fields with low intensity and intermediate frequency that exhibit anti-tumorigenic effects. While the effects of TTFields on glioblastoma cells have been studied previously, nothing is known about the influence of TTFields on MSCs. METHODS Single-cell RNA sequencing and immunofluorescence staining were employed to identify glioblastoma-associated MSCs in patient samples. Proliferation and clonogenic survival of human bone marrow-derived MSCs were assessed after TTFields in vitro. MSC' characteristic surface marker expression was determined using flow cytometry, while multi-lineage differentiation potential was examined with immunohistochemistry. Apoptosis was quantified based on caspase-3 and annexin-V/7-AAD levels in flow cytometry, and senescence was assessed with ß-galactosidase staining. MSCs' migratory potential was evaluated with Boyden chamber assays. RESULTS Single-cell RNA sequencing and immunofluorescence showed the presence of glioblastoma-associated MSCs in patient samples. TTFields significantly reduced proliferation and clonogenic survival of human bone marrow-derived MSCs by up to 60% and 90%, respectively. While the characteristic surface marker expression and differentiation capacity were intact after TTFields, treatment resulted in increased apoptosis and senescence. Furthermore, TTFields significantly reduced MSCs' migratory capacity. CONCLUSION We could demonstrate the presence of tumor-associated MSCs in glioblastoma patients, providing a rationale to study the impact of TTFields on MSCs. TTFields considerably increase apoptosis and senescence in MSCs, resulting in impaired survival and migration. The results provide a basis for further analyses on the role of MSCs in glioblastoma patients receiving TTFields.
Collapse
Affiliation(s)
- Maren Strack
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany
| | - Jan Kückelhaus
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Diebold
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Neurology and Medical Oncology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, German Red Cross Blood Service Baden- Württemberg- Hessen, Heidelberg University, Mannheim, Germany
| | - Peter E Huber
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
- Department of Radiation Oncology, University Hospital Center Heidelberg, Heidelberg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
- Department of Radiation Oncology, University of Leipzig Medical Center, Leipzig, Germany
- Comprehensive Cancer Center Central (CCCG) Germany, Partner Site Leipzig, Leipzig, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany.
- Department of Radiation Oncology, University of Leipzig Medical Center, Leipzig, Germany.
- Comprehensive Cancer Center Central (CCCG) Germany, Partner Site Leipzig, Leipzig, Germany.
| |
Collapse
|
9
|
Zheng M, Wang Y, Chen S, Suo Y, Yu J, Zhang X. Enhancing Electric Field Distribution in the Pancreas for Improved TTFields Therapy: A Computational Modeling Investigation. IEEE Trans Biomed Eng 2024; 71:2612-2619. [PMID: 38564342 DOI: 10.1109/tbme.2024.3383818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
BACKGROUND Tumor treating fields (TTFields) therapy has shown effectiveness in glioblastoma treatment and holds potential for other cancers. However, its application in pancreatic cancer and the distribution of electric fields in pancreas remain unexplored. This study aims to investigate the electric field distributions in pancreatic regions using different array configurations for TTFields therapy. METHODS Computational modelling was employed to simulate electric field distributions, and quantitative analysis was conducted. Human body impedance measurements were used to optimize the electric properties of the model. Various array configurations were examined to assess their impact on the electric field distributions. RESULTS The study revealed that well-positioned arrays, specifically the combination of 20-piece transducer arrays in anterior-posterior orientation and 13-piece transducer arrays in left-right orientation, consistently achieved electric fields exceeding the 1V/cm threshold in over 99.4% of the pancreas. Even with a reduced number of transducers (13 pieces for both orientations), sufficient electric field coverage was achieved, exceeding the threshold in over 92.9% of the pancreas. Additionally, different array placements within the same orientation were explored to address clinical challenges such as skin rash and patient anatomical variations. CONCLUSIONS This research lays the groundwork for understanding TTFields distribution within the abdomen, offering insights into optimizing array configurations for improved electric field delivery. These results offer promises of advancing TTFields therapy for pancreatic cancer towards clinical applications, and potentially enhancing treatment efficacy and patient outcomes.
Collapse
|
10
|
Liu L, Huang B, Lu Y, Zhao Y, Tang X, Shi Y. Interactions between electromagnetic radiation and biological systems. iScience 2024; 27:109201. [PMID: 38433903 PMCID: PMC10906530 DOI: 10.1016/j.isci.2024.109201] [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] [Indexed: 03/05/2024] Open
Abstract
Even though the bioeffects of electromagnetic radiation (EMR) have been extensively investigated during the past several decades, our understandings of the bioeffects of EMR and the mechanisms of the interactions between the biological systems and the EMRs are still far from satisfactory. In this article, we introduce and summarize the consensus, controversy, limitations, and unsolved issues. The published works have investigated the EMR effects on different biological systems including humans, animals, cells, and biochemical reactions. Alternative methodologies also include dielectric spectroscopy, detection of bioelectromagnetic emissions, and theoretical predictions. In many studies, the thermal effects of the EMR are not properly controlled or considered. The frequency of the EMR investigated is limited to the commonly used bands, particularly the frequencies of the power line and the wireless communications; far fewer studies were performed for other EMR frequencies. In addition, the bioeffects of the complex EM environment were rarely discussed. In summary, our understanding of the bioeffects of the EMR is quite restrictive and further investigations are needed to answer the unsolved questions.
Collapse
Affiliation(s)
- Lingyu Liu
- Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bing Huang
- Brain Function and Disease Laboratory, Department of Pharmacology, Shantou University Medical College, 22 Xin-Ling Road, Shantou 515041, China
| | - Yingxian Lu
- Westlake Laboratory of Life Sciences and Biomedicine, Xihu District, Hangzhou 310024, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Yanyu Zhao
- Westlake Laboratory of Life Sciences and Biomedicine, Xihu District, Hangzhou 310024, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Xiaping Tang
- Westlake Laboratory of Life Sciences and Biomedicine, Xihu District, Hangzhou 310024, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Yigong Shi
- Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Westlake Laboratory of Life Sciences and Biomedicine, Xihu District, Hangzhou 310024, Zhejiang Province, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
11
|
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
|
12
|
Tian W, Ning J, Chen L, Zeng Y, Shi Y, Xiao G, He S, Tanzhu G, Zhou R. Cost-effectiveness of tumor-treating fields plus standard therapy for advanced non-small cell lung cancer progressed after platinum-based therapy in the United States. Front Pharmacol 2024; 15:1333128. [PMID: 38375030 PMCID: PMC10875105 DOI: 10.3389/fphar.2024.1333128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
Background: Tumor treating fields (TTF) was first approved for treatment of glioblastoma. Recently, the LUNAR study demonstrated that TTF + standard therapy (ST) extended survival in patients with advanced non-small cell lung cancer (NSCLC). This primary objective of this study is to analyze the cost-effectiveness of this treatment from the United States healthcare payers' perspective. Methods: A 3-health-state Markov model was established to compare the cost-effectiveness of TTF + ST and that of ST alone. Clinical data were extracted from the LUNAR study, supplemented by additional cost and utility data obtained from publications or online sources. One-way sensitivity analysis, probabilistic sensitivity analysis, and scenario analysis were conducted. The willingness-to-pay (WTP) threshold per quality-adjusted life-years (QALYs) gained was set to $150,000. The main results include total costs, QALYs, incremental cost-effectiveness ratio (ICER) and incremental net monetary benefit (INMB). Subgroup analyses were conducted for two types of ST, including immune checkpoint inhibitor, and docetaxel. Results: During a 10-year time horizon, the costs of TTF + ST and ST alone were $431,207.0 and $128,125.9, and the QALYs were 1.809 and 1.124, respectively. The ICER of TTF + ST compared to ST was $442,732.7 per QALY, and the INMB was -$200,395.7 at the WTP threshold. The cost of TTF per month was the most influential factor in cost-effectiveness, and TTF + ST had a 0% probability of being cost-effective at the WTP threshold compared with ST alone. Conclusion: TTF + ST is not a cost-effective treatment for advanced NSCLC patients who progressed after platinum-based therapy from the perspective of the United States healthcare payers.
Collapse
Affiliation(s)
- Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Liu Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Zeng
- Changsha Stomatological Hospital, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Yin Shi
- Department of Pharmacy, Xiangya Hospital, Changsha, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuangshuang He
- Department of Radiation Oncology and Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Rehbein S, Possmayer AL, Bozkurt S, Lotsch C, Gerstmeier J, Burger M, Momma S, Maletzki C, Classen CF, Freiman TM, Dubinski D, Lamszus K, Stringer BW, Herold-Mende C, Münch C, Kögel D, Linder B. Molecular Determinants of Calcitriol Signaling and Sensitivity in Glioma Stem-like Cells. Cancers (Basel) 2023; 15:5249. [PMID: 37958423 PMCID: PMC10648216 DOI: 10.3390/cancers15215249] [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: 08/15/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Glioblastoma is the most common primary brain cancer in adults and represents one of the worst cancer diagnoses for patients. Suffering from a poor prognosis and limited treatment options, tumor recurrences are virtually inevitable. Additionally, treatment resistance is very common for this disease and worsens the prognosis. These and other factors are hypothesized to be largely due to the fact that glioblastoma cells are known to be able to obtain stem-like traits, thereby driving these phenotypes. Recently, we have shown that the in vitro and ex vivo treatment of glioblastoma stem-like cells with the hormonally active form of vitamin D3, calcitriol (1α,25(OH)2-vitamin D3) can block stemness in a subset of cell lines and reduce tumor growth. Here, we expanded our cell panel to over 40 different cultures and can show that, while half of the tested cell lines are sensitive, a quarter can be classified as high responders. Using genetic and proteomic analysis, we further determined that treatment success can be partially explained by specific polymorphism of the vitamin D3 receptor and that high responders display a proteome suggestive of blockade of stemness, as well as migratory potential.
Collapse
Affiliation(s)
- Sarah Rehbein
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, 60596 Frankfurt am Main, Germany; (S.R.); (A.-L.P.); (J.G.); (D.K.)
| | - Anna-Lena Possmayer
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, 60596 Frankfurt am Main, Germany; (S.R.); (A.-L.P.); (J.G.); (D.K.)
| | - Süleyman Bozkurt
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany; (S.B.); (C.M.)
| | - Catharina Lotsch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany (C.H.-M.)
| | - Julia Gerstmeier
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, 60596 Frankfurt am Main, Germany; (S.R.); (A.-L.P.); (J.G.); (D.K.)
| | - Michael Burger
- Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, 60596 Frankfurt am Main, Germany;
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, 60596 Frankfurt am Main, Germany;
| | - Claudia Maletzki
- Department of Medicine, Clinic III-Hematology, Oncology, Alliative Care Rostock, 18057 Rostock, Germany;
| | - Carl Friedrich Classen
- Division of Pediatric Oncology, Hematology and Palliative Medicine Section, Department of Pediatrics and Adolescent Medicine, University Medicine Rostock, 18057 Rostock, Germany;
| | - Thomas M. Freiman
- Department of Neurosurgery, University Hospital Rostock, 18057 Rostock, Germany; (T.M.F.); (D.D.)
| | - Daniel Dubinski
- Department of Neurosurgery, University Hospital Rostock, 18057 Rostock, Germany; (T.M.F.); (D.D.)
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg—Eppendorf, 20251 Hamburg, Germany;
| | - Brett W. Stringer
- College of Medicine and Public Health, Flinders University, Sturt Rd., Bedford Park, SA 5042, Australia;
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany (C.H.-M.)
| | - Christian Münch
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany; (S.B.); (C.M.)
| | - Donat Kögel
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, 60596 Frankfurt am Main, Germany; (S.R.); (A.-L.P.); (J.G.); (D.K.)
- German Cancer Consortium DKTK Partner Site Frankfurt/Main, 60590 Frankfurt am Main, Germany
- German Cancer Research Center DKFZ, 69120 Heidelberg, Germany
| | - Benedikt Linder
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, 60596 Frankfurt am Main, Germany; (S.R.); (A.-L.P.); (J.G.); (D.K.)
| |
Collapse
|
14
|
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
|
15
|
Leal T, Kotecha R, Ramlau R, Zhang L, Milanowski J, Cobo M, Roubec J, Petruzelka L, Havel L, Kalmadi S, Ward J, Andric Z, Berghmans T, Gerber DE, Kloecker G, Panikkar R, Aerts J, Delmonte A, Pless M, Greil R, Rolfo C, Akerley W, Eaton M, Iqbal M, Langer C. Tumor Treating Fields therapy with standard systemic therapy versus standard systemic therapy alone in metastatic non-small-cell lung cancer following progression on or after platinum-based therapy (LUNAR): a randomised, open-label, pivotal phase 3 study. Lancet Oncol 2023; 24:1002-1017. [PMID: 37657460 DOI: 10.1016/s1470-2045(23)00344-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Tumor Treating Fields (TTFields) are electric fields that disrupt processes critical for cancer cell survival, leading to immunogenic cell death and enhanced antitumour immune response. In preclinical models of non-small-cell lung cancer, TTFields amplified the effects of chemotherapy and immune checkpoint inhibitors. We report primary results from a pivotal study of TTFields therapy in metastatic non-small-cell lung cancer. METHODS This randomised, open-label, pivotal phase 3 study recruited patients at 130 sites in 19 countries. Participants were aged 22 years or older with metastatic non-small-cell lung cancer progressing on or after platinum-based therapy, with squamous or non-squamous histology and ECOG performance status of 2 or less. Previous platinum-based therapy was required, but no restriction was placed on the number or type of previous lines of systemic therapy. Participants were randomly assigned (1:1) to TTFields therapy and standard systemic therapy (investigator's choice of immune checkpoint inhibitor [nivolumab, pembrolizumab, or atezolizumab] or docetaxel) or standard therapy alone. Randomisation was performed centrally using variable blocked randomisation and an interactive voice-web response system, and was stratified by tumour histology, treatment, and region. Systemic therapies were dosed according to local practice guidelines. TTFields therapy (150 kHz) was delivered continuously to the thoracic region with the recommendation to achieve an average of at least 18 h/day device usage. The primary endpoint was overall survival in the intention-to-treat population. The safety population included all patients who received any study therapy and were analysed according to the actual treatment received. The study is registered with ClinicalTrials.gov, NCT02973789. FINDINGS Between Feb 13, 2017, and Nov 19, 2021, 276 patients were enrolled and randomly assigned to receive TTFields therapy with standard therapy (n=137) or standard therapy alone (n=139). The median age was 64 years (IQR 59-70), 178 (64%) were male and 98 (36%) were female, 156 (57%) had non-squamous non-small-cell lung cancer, and 87 (32%) had received a previous immune checkpoint inhibitor. Median follow-up was 10·6 months (IQR 6·1-33·7) for patients receiving TTFields therapy with standard therapy, and 9·5 months (0·1-32·1) for patients receiving standard therapy. Overall survival was significantly longer with TTFields therapy and standard therapy than with standard therapy alone (median 13·2 months [95% CI 10·3-15·5] vs 9·9 months [8·1-11·5]; hazard ratio [HR] 0·74 [95% CI 0·56-0·98]; p=0·035). In the safety population (n=267), serious adverse events of any cause were reported in 70 (53%) of 133 patients receiving TTFields therapy plus standard therapy and 51 (38%) of 134 patients receiving standard therapy alone. The most frequent grade 3-4 adverse events were leukopenia (37 [14%] of 267), pneumonia (28 [10%]), and anaemia (21 [8%]). TTFields therapy-related adverse events were reported in 95 (71%) of 133 patients; these were mostly (81 [85%]) grade 1-2 skin and subcutaneous tissue disorders. There were three deaths related to standard therapy (two due to infections and one due to pulmonary haemorrhage) and no deaths related to TTFields therapy. INTERPRETATION TTFields therapy added to standard therapy significantly improved overall survival compared with standard therapy alone in metastatic non-small-cell lung cancer after progression on platinum-based therapy without exacerbating systemic toxicities. These data suggest that TTFields therapy is efficacious in metastatic non-small-cell lung cancer and should be considered as a treatment option to manage the disease in this setting. FUNDING Novocure.
Collapse
Affiliation(s)
- Ticiana Leal
- Winship Cancer Institute at Emory University, Atlanta, GA, USA.
| | - Rupesh Kotecha
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Rodryg Ramlau
- Poznan University of Medical Sciences, Poznan, Poland
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
| | | | - Manuel Cobo
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, IBIMA, Málaga, Spain
| | - Jaromir Roubec
- Nemocnice AGEL Ostrava-Vítkovice, Ostrava, Czech Republic
| | | | | | | | - Jeffrey Ward
- Washington University School of Medicine, St Louis, MO, USA
| | - Zoran Andric
- University Clinical Hospital Centre Bezanijska Kosa, Belgrade, Serbia
| | - Thierry Berghmans
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - David E Gerber
- Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Joachim Aerts
- Department of Pulmonary Medicine, The Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Angelo Delmonte
- IRCCS Istituto Romagnolo per lo Studio dei Tumori Dino Amadori (IRST), Meldola, Italy
| | - Miklos Pless
- Kantonsspital Winterthur, Winterthur, Switzerland
| | - Richard Greil
- Salzburg Cancer Research Institute-Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Salzburg, Austria; Paracelsus Medical University Salzburg, Salzburg, Austria; Cancer Cluster, Salzburg, Austria
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute at Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Wallace Akerley
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Mussawar Iqbal
- College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Corey Langer
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
Ballo MT, Conlon P, Lavy-Shahaf G, Kinzel A, Vymazal J, Rulseh AM. Association of Tumor Treating Fields (TTFields) therapy with survival in newly diagnosed glioblastoma: a systematic review and meta-analysis. J Neurooncol 2023; 164:1-9. [PMID: 37493865 PMCID: PMC10462574 DOI: 10.1007/s11060-023-04348-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/16/2023] [Indexed: 07/27/2023]
Abstract
PURPOSE Tumor Treating Fields (TTFields) therapy, an electric field-based cancer treatment, became FDA-approved for patients with newly diagnosed glioblastoma (GBM) in 2015 based on the randomized controlled EF-14 study. Subsequent approvals worldwide and increased adoption over time have raised the question of whether a consistent survival benefit has been observed in the real-world setting, and whether device usage has played a role. METHODS We conducted a literature search to identify clinical studies evaluating overall survival (OS) in TTFields-treated patients. Comparative and single-cohort studies were analyzed. Survival curves were pooled using a distribution-free random-effects method. RESULTS Among nine studies, seven (N = 1430 patients) compared the addition of TTFields therapy to standard of care (SOC) chemoradiotherapy versus SOC alone and were included in a pooled analysis for OS. Meta-analysis of comparative studies indicated a significant improvement in OS for patients receiving TTFields and SOC versus SOC alone (HR: 0.63; 95% CI 0.53-0.75; p < 0.001). Among real-world post-approval studies, the pooled median OS was 22.6 months (95% CI 17.6-41.2) for TTFields-treated patients, and 17.4 months (95% CI 14.4-21.6) for those not receiving TTFields. Rates of gross total resection were generally higher in the real-world setting, irrespective of TTFields use. Furthermore, for patients included in studies reporting data on device usage (N = 1015), an average usage rate of ≥ 75% was consistently associated with prolonged survival (p < 0.001). CONCLUSIONS Meta-analysis of comparative TTFields studies suggests survival may be improved with the addition of TTFields to SOC for patients with newly diagnosed GBM.
Collapse
Affiliation(s)
- Matthew T Ballo
- Department of Radiation Oncology, West Cancer Center, Germantown, TN, USA
| | | | | | | | | | | |
Collapse
|
17
|
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
|
18
|
Kutuk T, Walker JM, Ballo MT, Cameron RB, Alvarez JB, Chawla S, Luk E, Behl D, Dal Pra A, Morganstein N, Refaat T, Sheybani A, Squillante C, Zhang J, Kotecha R. Multi-Institutional Patterns of Use of Tumor-Treating Fields for Patients with Malignant Pleural Mesothelioma. Curr Oncol 2023; 30:5195-5200. [PMID: 37366877 DOI: 10.3390/curroncol30060394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/28/2022] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
(1) Background: The objective of this analysis was to evaluate the device usage rates and patterns of use regarding Tumor-Treating Fields (TTFields) for patients with malignant pleural mesothelioma (MPM) throughout the US. (2) Methods: We evaluated de-identified data from 33 patients with MPM enrolled in FDA-required HDE protocols at 14 institutions across the US from September 2019 to March 2022. (3) Results: The median number of total TTFields usage days was 72 (range: 6-649 days), and the total treatment duration was 160 months for all patients. A low usage rate (defined as less than 6 h per day, 25%) was observed in 34 (21.2%) months. The median TTFields usage in the first 3 months was 12 h per day (range: 1.9-21.6 h), representing 50% (range: 8-90%) of the potential daily duration. The median TTFields usage after 3 months decreased to 9.1 h per day (range: 3.1-17 h), representing 38% (range: 13-71%) of the daily duration, and was lower than usage in the first 3 months (p = 0.01). (4) Conclusions: This study represents the first multicenter analysis of real-world TTFields usage based on usage patterns for MPM patients in clinical practice. The real-world usage level was lower than the suggested daily usage. Further initiatives and guidelines should be developed to evaluate the impact of this finding on tumor control.
Collapse
Affiliation(s)
- Tugce Kutuk
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
| | - Joshua M Walker
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Matthew T Ballo
- Department of Radiation Oncology, West Cancer Center & Research Institute, Memphis, TN 38138, USA
| | - Robert B Cameron
- Department of Thoracic Surgery, UCLA Health, Los Angeles, CA 90095, USA
| | - Jean Bustamante Alvarez
- Department of Thoracic Oncology, West Virginia University Healthcare, Morgantown, WV 26506, USA
| | - Sheema Chawla
- Department of Radiation Oncology, Rochester Regional Health, Rochester, NY 14621, USA
| | - Eric Luk
- Department of Medical Oncology, Ochsner Benson Cancer Center, Jefferson, LA 70121, USA
| | - Deepti Behl
- Department of Medical Oncology, Sutter Health-Sutter Cancer Center, Sacramento, CA 95816, USA
| | - Alan Dal Pra
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Neil Morganstein
- Department of Medical Oncology, Atlantic Health System, Morristown, NJ 07960, USA
| | - Tamer Refaat
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, USA
| | - Arshin Sheybani
- Department of Radiation Oncology, John Stoddard Cancer Center, Des Moines, IA 50309, USA
| | - Christian Squillante
- Department of Medical Oncology, Virginia Piper Cancer Institute, Minneapolis, MN 55404, USA
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 64154, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 64154, USA
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
| |
Collapse
|
19
|
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
|
20
|
Li H, Li JJ, Lu W, Yang J, Xia Y, Huang P. Targeting Mitochondrial IDH2 Enhances Antitumor Activity of Cisplatin in Lung Cancer via ROS-Mediated Mechanism. Biomedicines 2023; 11:biomedicines11020475. [PMID: 36831011 PMCID: PMC9953680 DOI: 10.3390/biomedicines11020475] [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: 01/04/2023] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Mitochondrial isocitrate dehydrogenase 2 (IDH2) is an important metabolic enzyme in the tricarboxylic acid cycle (TCA) cycle. Our previous study showed that high expression of wild-type IDH2 promotes the proliferation of lung cancer cells. This study aims to test the potential of targeting IDH2 as a therapeutic strategy to inhibit lung cancer in vitro and in vivo. First, we analyzed the available data from the databases gene expression omnibus (GEO) database to evaluate the clinical relevance of IDH2 expression in affecting lung cancer patient survival. We then generated a stable IDH2-knockdown lung cancer cell line using a lentivirus-based method for in vitro and in vivo study. Cell growth, apoptosis, cell viability, and colony formation assays were conducted to test the sensitivity of lung cancer cells with different IDH2 expression status to cisplatin or radiation treatment in vitro. For mechanistic study, Cellular oxygen consumption and extracellular acidification rates were measured using a Seahorse metabolic analyzer, and reactive oxygen species (ROS) generation was analyzed using flow cytometry. An animal study using a xenograft tumor model was performed to further evaluate the in vivo therapeutic effect on tumor growth. We found that high IDH2 expression was associated with poor survival in lung cancer patients undergoing chemotherapy. Inhibition of IDH2 significantly enhanced the anticancer activity of cisplatin and also increased the effect of radiation against lung cancer cells. IDH2 was upregulated in cisplatin-resistant lung cancer cells, which could be sensitized by targeted inhibition of IDH2. Mechanistic study showed that abrogation of IDH2 caused only minimal changes in oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) in lung cancer cells, but induced a significant increase in ROS, which rendered the cancer cells more sensitive to cisplatin. Pretreatment of lung cancer cells with the ROS scavenger N-acetyl-cysteine could partially rescue cells from the cytotoxic effect of cisplatin and IDH2 inhibition. Importantly, abrogation of IDH2 significantly increased the sensitivity of lung cancer cells to cisplatin in vivo.
Collapse
Affiliation(s)
- He Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Jiang-jiang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Jing Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yunfei Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
- Correspondence: (Y.X.); (P.H.); Tel.: +86-20-8734-3511 (P.H.)
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
- Metabolic Innovation Center, Sun Yat-sen University, Guangzhou 510060, China
- Correspondence: (Y.X.); (P.H.); Tel.: +86-20-8734-3511 (P.H.)
| |
Collapse
|
21
|
Current status of the preclinical evaluation of alternating electric fields as a form of cancer therapy. Bioelectrochemistry 2023; 149:108287. [DOI: 10.1016/j.bioelechem.2022.108287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
|
22
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524504. [PMID: 36789415 PMCID: PMC9928061 DOI: 10.1101/2023.01.18.524504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
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
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
Barsheshet Y, Voloshin T, Brant B, Cohen G, Koren L, Blatt R, Cahal S, Haj Khalil T, Zemer Tov E, Paz R, Klein-Goldberg A, Tempel-Brami C, Jacobovitch S, Volodin A, Kan T, Koltun B, David C, Haber A, Giladi M, Weinberg U, Palti Y. Tumor Treating Fields (TTFields) Concomitant with Immune Checkpoint Inhibitors Are Therapeutically Effective in Non-Small Cell Lung Cancer (NSCLC) In Vivo Model. Int J Mol Sci 2022; 23:ijms232214073. [PMID: 36430552 PMCID: PMC9696536 DOI: 10.3390/ijms232214073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/06/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Tumor Treating Fields (TTFields) are electric fields that exert physical forces to disrupt cellular processes critical for cancer cell viability and tumor progression. TTFields induce anti-mitotic effects through the disruption of the mitotic spindle and abnormal chromosome segregation, which trigger several forms of cell death, including immunogenic cell death (ICD). The efficacy of TTFields concomitant with anti-programmed death-1 (anti-PD-1) treatment was previously shown in vivo and is currently under clinical investigation. Here, the potential of TTFields concomitant with anti- PD-1/anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) or anti-programmed death-ligand 1 (anti-PD-L1) immune checkpoint inhibitors (ICI) to improve therapeutic efficacy was examined in lung tumor-bearing mice. Increased circulating levels of high mobility group box 1 protein (HMGB1) and elevated intratumoral levels of phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α) were found in the TTFields-treated mice, indicative of ICD induction. The concomitant application of TTFields and ICI led to a significant decrease in tumor volume as compared to all other groups. In addition, significant increases in the number of tumor-infiltrating immune cells, specifically cytotoxic T-cells, were observed in the TTFields plus anti-PD-1/anti-CTLA-4 or anti-PD-L1 groups. Correspondingly, cytotoxic T-cells isolated from these tumors showed higher levels of IFN-γ production. Collectively, these results suggest that TTFields have an immunoactivating role that may be leveraged for concomitant treatment with ICI to achieve better tumor control by enhancing antitumor immunity.
Collapse
|
27
|
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]
|
28
|
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
|
29
|
Tanzhu G, Chen L, Xiao G, Shi W, Peng H, Chen D, Zhou R. The schemes, mechanisms and molecular pathway changes of Tumor Treating Fields (TTFields) alone or in combination with radiotherapy and chemotherapy. Cell Death Discov 2022; 8:416. [PMID: 36220835 PMCID: PMC9553876 DOI: 10.1038/s41420-022-01206-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor Treating Fields (TTFields) is a physical therapy that uses moderate frequency (100-300 kHz) and low-intensity (1-3 V/cm) alternating electric fields to inhibit tumors. Currently, the Food and Drug Administration approves TTFields for treating recurrent or newly diagnosed glioblastoma (GBM) and malignant pleural mesothelioma (MPM). The classical mechanism of TTFields is mitotic inhibition by hindering the formation of tubulin and spindle. In addition, TTFields inhibits cell proliferation, invasion, migration and induces cell death, such as apoptosis, autophagy, pyroptosis, and cell cycle arrest. Meanwhile, it regulates immune function and changes the permeability of the nuclear membrane, cell membrane, and blood-brain barrier. Based on the current researches on TTFields in various tumors, this review comprehensively summarizes the in-vitro effects, changes in pathways and molecules corresponding to relevant parameters of TTFields (frequency, intensity, and duration). In addition, radiotherapy and chemotherapy are common tumor treatments. Thus, we also pay attention to the sequence and dose when TTFields combined with radiotherapy or chemotherapy. TTFields has inhibitory effects in a variety of tumors. The study of TTFields mechanism is conducive to subsequent research. How to combine common tumor therapy such as radiotherapy and chemotherapy to obtain the maximum benefit is also a problem that's worthy of our attention.
Collapse
Affiliation(s)
- Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Liu Chen
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Wen Shi
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Haiqin Peng
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P.R. China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, 410008, Changsha, China.
| |
Collapse
|
30
|
Salvador E, Kessler AF, Domröse D, Hörmann J, Schaeffer C, Giniunaite A, Burek M, Tempel-Brami C, Voloshin T, Volodin A, Zeidan A, Giladi M, Ernestus RI, Löhr M, Förster CY, Hagemann C. Tumor Treating Fields (TTFields) Reversibly Permeabilize the Blood-Brain Barrier In Vitro and In Vivo. Biomolecules 2022; 12:1348. [PMID: 36291557 PMCID: PMC9599321 DOI: 10.3390/biom12101348] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 10/02/2023] Open
Abstract
Despite the availability of numerous therapeutic substances that could potentially target CNS disorders, an inability of these agents to cross the restrictive blood-brain barrier (BBB) limits their clinical utility. Novel strategies to overcome the BBB are therefore needed to improve drug delivery. We report, for the first time, how Tumor Treating Fields (TTFields), approved for glioblastoma (GBM), affect the BBB's integrity and permeability. Here, we treated murine microvascular cerebellar endothelial cells (cerebEND) with 100-300 kHz TTFields for up to 72 h and analyzed the expression of barrier proteins by immunofluorescence staining and Western blot. In vivo, compounds normally unable to cross the BBB were traced in healthy rat brain following TTFields administration at 100 kHz. The effects were analyzed via MRI and immunohistochemical staining of tight-junction proteins. Furthermore, GBM tumor-bearing rats were treated with paclitaxel (PTX), a chemotherapeutic normally restricted by the BBB combined with TTFields at 100 kHz. The tumor volume was reduced with TTFields plus PTX, relative to either treatment alone. In vitro, we demonstrate that TTFields transiently disrupted BBB function at 100 kHz through a Rho kinase-mediated tight junction claudin-5 phosphorylation pathway. Altogether, if translated into clinical use, TTFields could represent a novel CNS drug delivery strategy.
Collapse
Affiliation(s)
- Ellaine Salvador
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Almuth F. Kessler
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Dominik Domröse
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Julia Hörmann
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Clara Schaeffer
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Aiste Giniunaite
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Malgorzata Burek
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, D-97080 Würzburg, Germany
| | | | | | | | | | | | - Ralf-Ingo Ernestus
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| | - Carola Y. Förster
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, D-97080 Würzburg, Germany
| | - Carsten Hagemann
- Department of Neurosurgery, Section Experimental Neurosurgery, University of Würzburg, D-97080 Würzburg, Germany
| |
Collapse
|
31
|
Le HT, Staelens M, Lazzari D, Chan G, Tuszyński JA. Real-Time Monitoring of the Effect of Tumour-Treating Fields on Cell Division Using Live-Cell Imaging. Cells 2022; 11:2712. [PMID: 36078119 PMCID: PMC9454843 DOI: 10.3390/cells11172712] [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: 04/30/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
The effects of electric fields (EFs) on various cell types have been thoroughly studied, and exhibit a well-known regulatory effect on cell processes, implicating their usage in several medical applications. While the specific effect exerted on cells is highly parameter-dependent, the majority of past research has focused primarily on low-frequency alternating fields (<1 kHz) and high-frequency fields (in the order of MHz). However, in recent years, low-intensity (1-3 V/cm) alternating EFs with intermediate frequencies (100-500 kHz) have been of topical interest as clinical treatments for cancerous tumours through their disruption of cell division and the mitotic spindle, which can lead to cell death. These aptly named tumour-treating fields (TTFields) have been approved by the FDA as a treatment modality for several cancers, such as malignant pleural mesothelioma and glioblastoma multiforme, demonstrating remarkable efficacy and a high safety profile. In this work, we report the results of in vitro experiments with HeLa and MCF-10A cells exposed to TTFields for 18 h, imaged in real time using live-cell imaging. Both studied cell lines were exposed to 100 kHz TTFields with a 1-1 duty cycle, which resulted in significant mitotic and cytokinetic arrest. In the experiments with HeLa cells, the effects of the TTFields' frequency (100 kHz vs. 200 kHz) and duty cycle (1-1 vs. 1-0) were also investigated. Notably, the anti-mitotic effect was stronger in the HeLa cells treated with 100 kHz TTFields. Additionally, it was found that single and two-directional TTFields (oriented orthogonally) exhibit a similar inhibitory effect on HeLa cell division. These results provide real-time evidence of the profound ability of TTFields to hinder the process of cell division by significantly delaying both the mitosis and cytokinesis phases of the cell cycle.
Collapse
Affiliation(s)
- Hoa T. Le
- Department of Medical Microbiology & Immunology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Michael Staelens
- Department of Physics, Faculty of Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Davide Lazzari
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, 10129 Turin, Italy
| | - Gordon Chan
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Jack A. Tuszyński
- Department of Physics, Faculty of Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
32
|
Tumor-Treating Fields in Glioblastomas: Past, Present, and Future. Cancers (Basel) 2022; 14:cancers14153669. [PMID: 35954334 PMCID: PMC9367615 DOI: 10.3390/cancers14153669] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Glioblastoma (GBM) is the most common malignant primary brain tumor. Although the standard of care, including maximal resection, concurrent radiotherapy with temozolomide (TMZ), and adjuvant TMZ, has largely improved the prognosis of these patients, the 5-year survival rate is still < 10%. Tumor-treating fields (TTFields), a noninvasive anticancer therapeutic modality, has been rising as a fourth treatment option for GBMs, as confirmed by recent milestone large-scale phase 3 randomized trials and subsequent real-world data, elongating patient overall survival from 16 months to 21 months. However, the mechanisms of antitumor efficacy, its clinical safety, and potential benefits when combined with other treatment modalities are far from completely elucidated. As an increasing number of studies have recently been published on this topic, we conducted this updated, comprehensive review to establish an objective understanding of the mechanism of action, efficacy, safety, clinical concerns, and future perspectives of TTFields. Abstract Tumor-treating fields (TTFields), a noninvasive and innovative therapeutic approach, has emerged as the fourth most effective treatment option for the management of glioblastomas (GBMs), the most deadly primary brain cancer. According to on recent milestone randomized trials and subsequent observational data, TTFields therapy leads to substantially prolonged patient survival and acceptable adverse events. Clinical trials are ongoing to further evaluate the safety and efficacy of TTFields in treating GBMs and its biological and radiological correlations. TTFields is administered by delivering low-intensity, intermediate-frequency, alternating electric fields to human GBM function through different mechanisms of action, including by disturbing cell mitosis, delaying DNA repair, enhancing autophagy, inhibiting cell metabolism and angiogenesis, and limiting cancer cell migration. The abilities of TTFields to strengthen intratumoral antitumor immunity, increase the permeability of the cell membrane and the blood–brain barrier, and disrupt DNA-damage-repair processes make it a promising therapy when combined with conventional treatment modalities. However, the overall acceptance of TTFields in real-world clinical practice is still low. Given that increasing studies on this promising topic have been published recently, we conducted this updated review on the past, present, and future of TTFields in GBMs.
Collapse
|
33
|
Yang Y, Hu X, Liu Y, Ouyang B, Zhang J, Jin H, Yu Z, Liu R, Li Z, Jiang L, Lin X, Xu B. An implantable ultrasound-powered device for the treatment of brain cancer using electromagnetic fields. SCIENCE ADVANCES 2022; 8:eabm5023. [PMID: 35867783 PMCID: PMC9307245 DOI: 10.1126/sciadv.abm5023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Brain tumors have been proved challenging to treat. Here, we present a promising alternative by developing an implantable ultrasound-powered tumor treating device (UP-TTD) that electromagnetically disrupts the rapid division of cancer cells without any adverse effects on normal neurons, thereby safely inhibiting brain cancer recurrence. In vitro and in vivo experiments confirmed the significant therapeutic effect of the UP-TTD, with ~58% inhibition on growth rate of clinical tumor cells and ~78% reduction of cancer area in tumor-bearing rats. This UP-TTD is wireless ultrasound-powered, chip-sized, lightweight, and easy to operate on complex surfaces, with a largely boosting therapeutic efficiency and reducing energy consumption. Meanwhile, various treatment parameters could be tuned from the UP-TTD without increasing its size or adding circuits on the integrated chip. The tuning process was simulated and discussed, showing an excellent agreement with the experimental data. The encouraging results of the UP-TTD raise the possibility of a new modality for brain cancer treatment.
Collapse
Affiliation(s)
- Yilin Yang
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Xiaoping Hu
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Yuxin Liu
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Bin Ouyang
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Jiaxi Zhang
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Huawei Jin
- The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd., Yuexiu District, Guangzhou, Guangdong 510080, P.R. China
| | - Zhenhua Yu
- The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Rd., Yuexiu District, Guangzhou, Guangdong 510080, P.R. China
| | - Ruiwei Liu
- School of Naval Architecture and Ocean Engineering, Guangzhou Maritime University, 101 Hongshan 3rd Road, Huangpu District, Guangzhou, Guangdong 510725, P.R. China
| | - Zhe Li
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Lelun Jiang
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Xudong Lin
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
| | - Bingzhe Xu
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen Campus, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, P.R. China
- Corresponding author.
| |
Collapse
|
34
|
Staelens M, Di Gregorio E, Kalra AP, Le HT, Hosseinkhah N, Karimpoor M, Lim L, Tuszyński JA. Near-Infrared Photobiomodulation of Living Cells, Tubulin, and Microtubules In Vitro. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:871196. [PMID: 35600165 PMCID: PMC9115106 DOI: 10.3389/fmedt.2022.871196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
We report the results of experimental investigations involving photobiomodulation (PBM) of living cells, tubulin, and microtubules in buffer solutions exposed to near-infrared (NIR) light emitted from an 810 nm LED with a power density of 25 mW/cm2 pulsed at a frequency of 10 Hz. In the first group of experiments, we measured changes in the alternating current (AC) ionic conductivity in the 50–100 kHz range of HeLa and U251 cancer cell lines as living cells exposed to PBM for 60 min, and an increased resistance compared to the control cells was observed. In the second group of experiments, we investigated the stability and polymerization of microtubules under exposure to PBM. The protein buffer solution used was a mixture of Britton-Robinson buffer (BRB aka PEM) and microtubule cushion buffer. Exposure of Taxol-stabilized microtubules (~2 μM tubulin) to the LED for 120 min resulted in gradual disassembly of microtubules observed in fluorescence microscopy images. These results were compared to controls where microtubules remained stable. In the third group of experiments, we performed turbidity measurements throughout the tubulin polymerization process to quantify the rate and amount of polymerization for PBM-exposed tubulin vs. unexposed tubulin samples, using tubulin resuspended to final concentrations of ~ 22.7 μM and ~ 45.5 μM in the same buffer solution as before. Compared to the unexposed control samples, absorbance measurement results demonstrated a slower rate and reduced overall amount of polymerization in the less concentrated tubulin samples exposed to PBM for 30 min with the parameters mentioned above. Paradoxically, the opposite effect was observed in the 45.5 μM tubulin samples, demonstrating a remarkable increase in the polymerization rates and total polymer mass achieved after exposure to PBM. These results on the effects of PBM on living cells, tubulin, and microtubules are novel, further validating the modulating effects of PBM and contributing to designing more effective PBM parameters. Finally, potential consequences for the use of PBM in the context of neurodegenerative diseases are discussed.
Collapse
Affiliation(s)
- Michael Staelens
- Department of Physics, University of Alberta, Edmonton, AB, Canada
| | | | - Aarat P. Kalra
- Scholes Lab, Department of Chemistry, Princeton University, Princeton, NJ, United States
| | - Hoa T. Le
- Department of Physics, University of Alberta, Edmonton, AB, Canada
| | | | | | - Lew Lim
- Vielight Inc., Toronto, ON, Canada
| | - Jack A. Tuszyński
- Department of Physics, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Jack A. Tuszyński
| |
Collapse
|
35
|
Skull modulated strategies to intensify tumor treating fields on brain tumor: a finite element study. Biomech Model Mechanobiol 2022; 21:1133-1144. [PMID: 35477828 DOI: 10.1007/s10237-022-01580-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 03/25/2022] [Indexed: 11/02/2022]
Abstract
Tumor treating fields (TTFields) are a breakthrough in treating glioblastoma (GBM), whereas the intensity cannot be further enhanced, due to the limitation of scalp lesions. Skull remodeling (SR) surgery can elevate the treatment dose of TTFields in the intracranial foci. This study was aimed at exploring the characteristics of the skull modulated strategies toward TTFields augmentation. The simplified multiple-tissue-layer model (MTL) and realistic head (RH) model were reconstructed through finite element methods (FEM), to simulate the remodeling of the skull, which included skull drilling, thinning, and cranioplasty with PEEK, titanium, cerebrospinal fluid (CSF), connective tissue and autologous bone. Skull thinning could enhance the intensity of TTFields in the brain tumor, with a 10% of increase in average peritumoral intensity (API) by every 1 cm decrease in skull thickness. Cranioplasty with titanium accompanied the most enhancement of TTFields in the MTL model, but CSF was superior in TTFields enhancement when simulated in the RH model. Besides, API increased nonlinearly with the expansion of drilled burr holes. In comparison with the single drill replaced by titanium, nine burr holes could reach 96.98% of enhancement in API, but it could only reach 63.08% of enhancement under craniectomy of nine times skull defect area. Skull thinning and drilling could enhance API, which was correlated with the number and area of skull drilling. Cranioplasty with highly conductive material could also augment API, but might not provide clinical benefits as expected.
Collapse
|
36
|
Feasibility of Tumor Treating Fields with Pemetrexed and Platinum-Based Chemotherapy for Unresectable Malignant Pleural Mesothelioma: Single-Center, Real-World Data. Cancers (Basel) 2022; 14:cancers14082020. [PMID: 35454925 PMCID: PMC9032984 DOI: 10.3390/cancers14082020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Management of malignant pleural mesothelioma (MPM) is challenging as patients frequently present with unresectable disease and the response rates with systemic therapy alone remain low. Given the paucity of effective therapies for MPM, Tumor Treating Fields (TTFields) therapy was made available for use under an FDA-approved Humanitarian Device Exemption (HDE) protocol in 2019, but no real-world data beyond the initial trial have been published to date. We reviewed our retrospective series of five patients diagnosed with MPM and treated with TTFields with pemetrexed and platinum-based chemotherapy. This therapy resulted in a modest disease-stabilization rate with no significant device-related major toxicities. However, we observed universal low-grade skin toxicities related to the device which required medical management and self-discontinuation in 2/5 patients. We also observed lower device usage, compared to the STELLAR trial. Both of these represent opportunities for developing improved management guidelines and efforts to improve patient compliance. Abstract Purpose: The objectives of this study were to evaluate the implementation, device usage rates, clinical outcomes, and treatment-related toxicities associated with TTFields and pemetrexed plus platinum-based chemotherapy in patients with unresectable MPM, outside the initial trial results. Methods: Consecutive patients with unresectable MPM were enrolled onto an FDA-required HDE protocol from 2019 to 2021. All patients were treated with a protocol-defined regimen of continuous TTFields (150 kHz) and pemetrexed plus platinum-based chemotherapy. Results: Five patients with unresectable MPM were enrolled. The median number of 4-week TTFields cycles was 5 (range: 2–7 cycles). Median TTFields device usage in the first 3 months was 12.5 h per day (range: 5–16.8 h), representing 52% (21–70%) of the potential daily duration. The median follow-up was 5.4 months (range: 1.1–20.9 months). Treatment-related dermatitis was the only side effect associated with TTFields and was reported as grade 1–2 in all patients; no patient had grade 3+ device-related toxicities. Conclusions: This study represents the first results of real-world implementation of TTFields for MPM. In comparison to the initial clinical trial (STELLAR), compliance rates were lower, although skin-related toxicities appeared similar. Further initiatives and guidelines should be developed to manage treatment-related dermatitis and improve device usage.
Collapse
|
37
|
Jones TH, Song JW, Abushahin L. Tumor treating fields: An emerging treatment modality for thoracic and abdominal cavity cancers. Transl Oncol 2022; 15:101296. [PMID: 34847422 PMCID: PMC8633677 DOI: 10.1016/j.tranon.2021.101296] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023] Open
Abstract
Tumor treating fields (TTFields)-an intermediate-frequency, electric field therapy-has emerged as a promising alternative therapy for the treatment of solid cancers. Since the first publication describing the anticancer effects of TTFields in 2004 there have been numerous follow-up studies by other groups, either to confirm the efficacy of TTFields or to study the primary mechanism of interaction. The overwhelming conclusion from these in vitro studies is that TTFields reduce the viability of aggressively replicating cell lines. However, there is still speculation as to the primary mechanism for this effect; moreover, observations both in vitro and in vivo of inhibited migration and metastases have been made, which may be unrelated to the originally proposed hypothesis of replication stress. Adding to this, the in vivo environment is much more complex spatially, structurally, and involves intricate networks of cell signaling, all of which could change the efficacy of TTFields in the same way pharmaceutical interventions often struggle transitioning in vivo. Despite this, TTFields have shown promise in clinical practice on multiple cancer types, which begs the question: has the primary mechanism carried over from in vitro to in vivo or are there new mechanisms at play? The goal of this review is to highlight the current proposed mechanism of action of TTFields based primarily on in vitro experiments and animal models, provide a summary of the clinical efficacy of TTFields, and finally, propose future directions of research to identify all possible mechanisms in vivo utilizing novel tumor-on-a-chip platforms.
Collapse
Affiliation(s)
- Travis H Jones
- Department of Mechanical and Aerospace Engineering, The Ohio State University, 201W. 19th Avenue, E406 Scott Laboratory, Columbus, OH 43210, United States; Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, 1800 Canon Drive, 1300G, Columbus, OH 43210, United States
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, 201W. 19th Avenue, E406 Scott Laboratory, Columbus, OH 43210, United States; Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, United States.
| | - Laith Abushahin
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, 1800 Canon Drive, 1300G, Columbus, OH 43210, United States; Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, United States.
| |
Collapse
|
38
|
Hong P, Kudulaiti N, Wu S, Nie J, Zhuang D. Tumor treating fields: a comprehensive overview of the underlying molecular mechanism. Expert Rev Mol Diagn 2021; 22:19-28. [PMID: 34883030 DOI: 10.1080/14737159.2022.2017283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION As a novel treatment modality, tumor treating fields (TTFields) exert low-intensity, medium-frequency electric fields on tumor cells. TTFields' effectiveness and safety have been demonstrated clinically and in the real world for treating glioblastoma, the most common and aggressive primary central nervous system tumor. TTFields therapy has also been approved for the management of malignant mesothelioma, and clinical trials are ongoing for NSCLC, gastric cancer, pancreatic cancer, and other solid tumors. AREAS COVERED This article comprehensively reviews the currently described evidence of TTFields' mechanism of action. TTFields' most evident therapeutic effect is to induce cell death by disrupting mitosis. Moreover, evidence suggests at additional mechanistic complexity, such as delayed DNA repair and heightened DNA replication stress, reversible increase in cell membrane and blood-brain barrier permeability, induction of immune response, and so on. EXPERT OPINION TTFields therapy has been arising as the fourth anti-tumor treatment besides surgery, radiotherapy, and antineoplastic agents in recent years. However, the precise molecular mechanisms underlying the effects of TTFields are not fully understood and some concepts remain controversial. An in-depth understanding of TTFields' effects on tumor cell and tumor microenvironment would be crucial for informing research aimed at further optimizing TTFields' efficacy and developing new combination therapies for clinical applications.
Collapse
Affiliation(s)
- Pengjie Hong
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Nijiati Kudulaiti
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Shuai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Jingtao Nie
- Zai Lab Trading (Shanghai) Co., Ltd., Shanghai, China
| | - Dongxiao Zhuang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| |
Collapse
|
39
|
Arvind R, Chandana SR, Borad MJ, Pennington D, Mody K, Babiker H. Tumor-Treating Fields: A fourth modality in cancer treatment, new practice updates. Crit Rev Oncol Hematol 2021; 168:103535. [PMID: 34808377 DOI: 10.1016/j.critrevonc.2021.103535] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/12/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022] Open
Abstract
Although major innovations in treatment are advancing, cancer persists as one of the leading causes of mortality. With the rising incidence of cancer and as we treat them, patients incur short term and long-term toxicities of current traditional therapies, including chemotherapy. This imposes a significant physical, emotional, and financial burden among patients, which affects their quality of life. Tumor-Treating Fields (TTFields) is a novel innovative new treatment modality that utilizes alternating electric fields at specific intermediate frequencies to diminish tumor growth by inhibiting mitosis and thus proliferation of malignant cells. The distinguishing feature of this new treatment modality is that it is noninvasive and tolerable. In fact, TTFields is currently FDA approved for the treatment of glioblastoma multiforme (GBM) as well as malignant pleural mesothelioma (MPM). Recently, TTFields have also been found to affect immunogenic cell death resulting in stronger anti-neoplastic effects. In this review, we discuss the mechanism of action of TTFields, the plethora of clinical trials being conducted in patients with GBM, pancreatic adenocarcinoma, ovarian cancer, non-small-cell-lung-cancer (NSCLC), brain metastasis from NSCLC, and MPM and toxicity profile.
Collapse
Affiliation(s)
- Rhea Arvind
- University of Arizona, College of Science, Tucson, AZ, USA
| | - Sreenivasa R Chandana
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, Phase I Program, START Midwest, Grand Rapids, MI, USA
| | - Mitesh J Borad
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Danniel Pennington
- University of Arizona Cancer Center, Clinical Trials Office, Tucson, AZ, USA
| | - Kabir Mody
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Hani Babiker
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
40
|
Hernández-Bule ML, Martínez MA, Trillo MÁ, Martínez L, Toledano-Macías E, Úbeda A. Response of human cancer cells to simultaneous treatment with sorafenib and radiofrequency current. Oncol Lett 2021; 22:807. [PMID: 34630714 PMCID: PMC8488331 DOI: 10.3892/ol.2021.13068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/24/2021] [Indexed: 01/16/2023] Open
Abstract
Due to their alleged analgesic, anti-inflammatory and tissue regenerative effects, capacitive-resistive electrothermal therapy (CRET), which is based on non-invasive exposure to radiofrequency (RF) currents, is often applied to chemotherapeutically treated patients with cancer. Our previous studies have demonstrated that subthermal CRET currents can elicit a number of cell responses, including anti-proliferative effects, in the human liver cancer cell line HepG2. Such effects involve significant changes in the regulation of proteins involved in MAPK signaling pathways, which are also implicated in the cancer cell response to standard anticancer drugs such as sorafenib. This overlap in response pathways may lead to competitive, neutralizing or blocking interactions between the electrical and chemical treatments, thus raising questions on the advisability of CRET treatment for their analgesic, anti-inflammatory or other purposes in patients undergoing chemotherapy. The present study analyzed the effects of simultaneous treatment with sorafenib and 448-kHz, subthermal CRET current on the proliferation and viability of HepG2 cell cultures. Cell viability was assessed through Trypan blue or XTT assays, while flow cytometry was applied for cell cycle and apoptosis analysis. The expression of proteins involved in cell proliferation were assessed by immunoblotting and immunofluorescence. The results revealed no evidence to suggest that the electrical treatment counteracted or neutralized the cellular response to sorafenib at the different conditions evaluated. Furthermore, at the standard pharmacological sorafenib concentration, 5 µM, the combined treatment elicited an anti-proliferative response significantly stronger than that induced by each of the treatments when applied separately in HepG2 cells. These data do not support the hypothesis that CRET exposure may inhibit or diminish the effects of a chemotherapeutic drug used in cancer treatment, and highlights the requirement for further investigation into the cell response to the combined action of electrical and chemical treatments.
Collapse
Affiliation(s)
| | - María Antonia Martínez
- Bioelectromagnetism-Research Service, Ramón y Cajal University Hospital, IRYCIS, Madrid 28034, Spain
| | - María Ángeles Trillo
- Bioelectromagnetism-Research Service, Ramón y Cajal University Hospital, IRYCIS, Madrid 28034, Spain
| | - Lidia Martínez
- Bioelectromagnetism-Research Service, Ramón y Cajal University Hospital, IRYCIS, Madrid 28034, Spain
| | - Elena Toledano-Macías
- Bioelectromagnetism-Research Service, Ramón y Cajal University Hospital, IRYCIS, Madrid 28034, Spain
| | - Alejandro Úbeda
- Bioelectromagnetism-Research Service, Ramón y Cajal University Hospital, IRYCIS, Madrid 28034, Spain
| |
Collapse
|
41
|
Mahgoub E, Hussain A, Sharifi M, Falahati M, Marei HE, Hasan A. The therapeutic effects of tumor treating fields on cancer and noncancerous cells. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
42
|
Mumblat H, Martinez-Conde A, Braten O, Munster M, Dor-On E, Schneiderman RS, Porat Y, Voloshin T, Davidi S, Blatt R, Shteingauz A, Tempel-Brami C, Zeevi E, Lajterer C, Shmueli Y, Danilov S, Haber A, Giladi M, Weinberg U, Kinzel A, Palti Y. Tumor Treating Fields (TTFields) downregulate the Fanconi Anemia-BRCA pathway and increase the efficacy of chemotherapy in malignant pleural mesothelioma preclinical models. Lung Cancer 2021; 160:99-110. [PMID: 34482104 DOI: 10.1016/j.lungcan.2021.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields with antimitotic effects on cancerous cells. TTFields concomitant with pemetrexed and a platinum agent are approved in the US and EU as first line therapy for unresectable, locally advanced or metastatic malignant pleural mesothelioma (MPM). The goal of the current study was to characterize the mechanism of action of TTFields in MPM cell lines and animal models. METHODS Human MPM cell lines MSTO-211H and NCI-H2052 were treated with TTFields to determine the frequency that elicits maximal cytotoxicity. The effect of TTFields on DNA damage and repair, and the cytotoxic effect of TTFields in combination with cisplatin and/or pemetrexed were examined. Efficacy of TTFields concomitant with cisplatin and pemetrexed was evaluated in orthotopic IL-45 and subcutaneous RN5 murine models. RESULTS TTFields at a frequency of 150 kHz demonstrated the highest cytotoxicity to MPM cells. Application of 150 kHz TTFields resulted in increased formation of DNA double strand breaks, elevated expression of DNA damage induced cell cycle arrest proteins, and reduced expression of Fanconi Anemia (FA)-BRCA DNA repair pathway proteins. Co-treatment of TTFields with cisplatin or pemetrexed significantly increased treatment efficacy versus each modality alone, with additivity and synergy exhibited by the TTFields-pemetrexed and TTFields-cisplatin combinations, respectively. In animal models, tumor volume was significantly lower for the TTFields-cisplatin-pemetrexed combination compared to control, accompanied by increased DNA damage within the tumor. CONCLUSION This research demonstrated that the efficacy of TTFields for the treatment of MPM is associated with reduced expression of FA-BRCA pathway proteins and increased DNA damage. This mechanism of action is consistent with the observed synergism for TTFields-cisplatin vs additivity for TTFields-pemetrexed, as cisplatin-induced DNA damage is repaired via the FA-BRCA pathway.
Collapse
|
43
|
Gerstmeier J, Possmayer AL, Bozkurt S, Hoffmann ME, Dikic I, Herold-Mende C, Burger MC, Münch C, Kögel D, Linder B. Calcitriol Promotes Differentiation of Glioma Stem-Like Cells and Increases Their Susceptibility to Temozolomide. Cancers (Basel) 2021; 13:cancers13143577. [PMID: 34298790 PMCID: PMC8303292 DOI: 10.3390/cancers13143577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 07/13/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Cancer cells with a stem-like phenotype that are thought to be highly tumorigenic are commonly described in glioblastoma, the most common primary adult brain cancer. This phenotype comprises high self-renewal capacity and resistance against chemotherapy and radiation therapy, thereby promoting tumor progression and disease relapse. Here, we show that calcitriol, the hormonally active form of the “sun hormone” vitamin D3, effectively suppresses stemness properties in glioblastoma stem-like cells (GSCs), supporting the hypothesis that calcitriol sensitizes them to additional chemotherapy. Indeed, a physiological organotypic brain slice model was used to monitor tumor growth of GSCs, and the effectiveness of combined treatment with temozolomide, the current standard-of-care, and calcitriol was proven. These findings indicate that further research on applying calcitriol, a well-known and safe drug, as a potential adjuvant therapy for glioblastoma is both justified and necessary. Abstract Glioblastoma (GBM) is the most common and most aggressive primary brain tumor, with a very high rate of recurrence and a median survival of 15 months after diagnosis. Abundant evidence suggests that a certain sub-population of cancer cells harbors a stem-like phenotype and is likely responsible for disease recurrence, treatment resistance and potentially even for the infiltrative growth of GBM. GBM incidence has been negatively correlated with the serum levels of 25-hydroxy-vitamin D3, while the low pH within tumors has been shown to promote the expression of the vitamin D3-degrading enzyme 24-hydroxylase, encoded by the CYP24A1 gene. Therefore, we hypothesized that calcitriol can specifically target stem-like glioblastoma cells and induce their differentiation. Here, we show, using in vitro limiting dilution assays, quantitative real-time PCR, quantitative proteomics and ex vivo adult organotypic brain slice transplantation cultures, that therapeutic doses of calcitriol, the hormonally active form of vitamin D3, reduce stemness to varying extents in a panel of investigated GSC lines, and that it effectively hinders tumor growth of responding GSCs ex vivo. We further show that calcitriol synergizes with Temozolomide ex vivo to completely eliminate some GSC tumors. These findings indicate that calcitriol carries potential as an adjuvant therapy for a subgroup of GBM patients and should be analyzed in more detail in follow-up studies.
Collapse
Affiliation(s)
- Julia Gerstmeier
- Neuroscience Center, Experimental Neurosurgery, Department of Neurosurgery, Goethe University, 60590 Frankfurt am Main, Germany; (J.G.); (A.-L.P.); (D.K.)
| | - Anna-Lena Possmayer
- Neuroscience Center, Experimental Neurosurgery, Department of Neurosurgery, Goethe University, 60590 Frankfurt am Main, Germany; (J.G.); (A.-L.P.); (D.K.)
| | - Süleyman Bozkurt
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany; (S.B.); (M.E.H.); (I.D.); (C.M.)
| | - Marina E. Hoffmann
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany; (S.B.); (M.E.H.); (I.D.); (C.M.)
| | - Ivan Dikic
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany; (S.B.); (M.E.H.); (I.D.); (C.M.)
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany;
| | - Michael C. Burger
- Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, 60528 Frankfurt am Main, Germany;
| | - Christian Münch
- Faculty of Medicine, Institute of Biochemistry II, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany; (S.B.); (M.E.H.); (I.D.); (C.M.)
| | - Donat Kögel
- Neuroscience Center, Experimental Neurosurgery, Department of Neurosurgery, Goethe University, 60590 Frankfurt am Main, Germany; (J.G.); (A.-L.P.); (D.K.)
- German Cancer Consortium DKTK Partner Site Frankfurt/Main, 60590 Frankfurt am Main, Germany
- German Cancer Research Center DKFZ, 69120 Heidelberg, Germany
| | - Benedikt Linder
- Neuroscience Center, Experimental Neurosurgery, Department of Neurosurgery, Goethe University, 60590 Frankfurt am Main, Germany; (J.G.); (A.-L.P.); (D.K.)
- Correspondence: ; Tel.: +49-69-6301-6930
| |
Collapse
|
44
|
Therapy of pancreatic cancer with alternating electric fields: Limitations of the method. Bioelectrochemistry 2021; 141:107881. [PMID: 34245959 DOI: 10.1016/j.bioelechem.2021.107881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/08/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with a poor prognosis. More effective treatment options are urgently needed. The use of physical and weak alternating electric fields (TTFields) can inhibit cell division of PDAC carcinoma and is currently being investigated in clinical trials. Here, we analyzed this new physical treatment under non-ideal conditions such as may occur during patient treatment. Three established human PDAC cell lines BxPC-3, gemcitabine-resistant BxPC-3 (BxGem), AsPC-1, and a non-malignant primary pancreatic cell line CRL-4023 were treated with TTFields in vitro. MTT assays, electrical impedance measurement, cell staining with Annexin V/7AAD followed by FACS analysis, digital image analysis and immunohistochemistry were performed. Treatment with TTFields smaller than 0.7 V/cm and field lines in the direction of mitotic spindle orientation significantly inhibited proliferation of all PDAC cells at 150 kHz, but significantly increased viability of AsPC-1 cells at all frequencies between 100 kHz and 300 kHz and that of BxPC-3 and BxGem cells at 250 kHz. Apoptosis or necrosis were not induced. Non-malignant CRL-4023 cells were not affected at 150 kHz. TTFields damaged PDAC cell lines but even favored their viability at very weak field strength and unfavorable frequency or inadequate field direction.
Collapse
|
45
|
Blatt R, Davidi S, Munster M, Shteingauz A, Cahal S, Zeidan A, Marciano T, Bomzon Z, Haber A, Giladi M, Weinberg U, Kinzel A, Palti Y. In Vivo Safety of Tumor Treating Fields (TTFields) Applied to the Torso. Front Oncol 2021; 11:670809. [PMID: 34249709 PMCID: PMC8264759 DOI: 10.3389/fonc.2021.670809] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022] Open
Abstract
Background Tumor Treating Fields (TTFields) therapy is a non-invasive, loco-regional, anti-mitotic treatment modality that targets rapidly dividing cancerous cells, utilizing low intensity, alternating electric fields at cancer-cell-type specific frequencies. TTFields therapy is approved for the treatment of newly diagnosed and recurrent glioblastoma (GBM) in the US, Europe, Israel, Japan, and China. The favorable safety profile of TTFields in patients with GBM is partially attributed to the low rate of mitotic events in normal, quiescent brain cells. However, specific safety evaluations are warranted at locations with known high rates of cellular proliferation, such as the torso, which is a primary site of several of the most aggressive malignant tumors. Methods The safety of delivering TTFields to the torso of healthy rats at 150 or 200 kHz, which were previously identified as optimal frequencies for treating multiple torso cancers, was investigated. Throughout 2 weeks of TTFields application, animals underwent daily clinical examinations, and at treatment cessation blood samples and internal organs were examined. Computer simulations were performed to verify that the targeted internal organs of the torso were receiving TTFields at therapeutic intensities (≥ 1 V/cm root mean square, RMS). Results No treatment-related mortality was observed. Furthermore, no significant differences were observed between the TTFields-treated and control animals for all examined safety parameters: activity level, food and water intake, stools, motor neurological status, respiration, weight, complete blood count, blood biochemistry, and pathological findings of internal organs. TTFields intensities of 1 to 2.5 V/cm RMS were confirmed for internal organs within the target region. Conclusions This research demonstrates the safety of therapeutic level TTFields at frequencies of 150 and 200 kHz when applied as monotherapy to the torso of healthy rats.
Collapse
|
46
|
Aguilar AA, Ho MC, Chang E, Carlson KW, Natarajan A, Marciano T, Bomzon Z, Patel CB. Permeabilizing Cell Membranes with Electric Fields. Cancers (Basel) 2021; 13:2283. [PMID: 34068775 PMCID: PMC8126200 DOI: 10.3390/cancers13092283] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/29/2022] Open
Abstract
The biological impact of exogenous, alternating electric fields (AEFs) and direct-current electric fields has a long history of study, ranging from effects on embryonic development to influences on wound healing. In this article, we focus on the application of electric fields for the treatment of cancers. In particular, we outline the clinical impact of tumor treating fields (TTFields), a form of AEFs, on the treatment of cancers such as glioblastoma and mesothelioma. We provide an overview of the standard mechanism of action of TTFields, namely, the capability for AEFs (e.g., TTFields) to disrupt the formation and segregation of the mitotic spindle in actively dividing cells. Though this standard mechanism explains a large part of TTFields' action, it is by no means complete. The standard theory does not account for exogenously applied AEFs' influence directly upon DNA nor upon their capacity to alter the functionality and permeability of cancer cell membranes. This review summarizes the current literature to provide a more comprehensive understanding of AEFs' actions on cell membranes. It gives an overview of three mechanistic models that may explain the more recent observations into AEFs' effects: the voltage-gated ion channel, bioelectrorheological, and electroporation models. Inconsistencies were noted in both effective frequency range and field strength between TTFields versus all three proposed models. We addressed these discrepancies through theoretical investigations into the inhomogeneities of electric fields on cellular membranes as a function of disease state, external microenvironment, and tissue or cellular organization. Lastly, future experimental strategies to validate these findings are outlined. Clinical benefits are inevitably forthcoming.
Collapse
Affiliation(s)
- Alondra A. Aguilar
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Michelle C. Ho
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Edwin Chang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Kristen W. Carlson
- Beth Israel Deaconess Medical Center, Department of Neurosurgery, Harvard Medical School, Boston, MA 02215, USA;
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
| | - Tal Marciano
- Novocure, Ltd., 31905 Haifa, Israel; (T.M.); (Z.B.)
| | - Ze’ev Bomzon
- Novocure, Ltd., 31905 Haifa, Israel; (T.M.); (Z.B.)
| | - Chirag B. Patel
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; (A.A.A.); (M.C.H.); (E.C.); (A.N.)
- Department of Neurology & Neurological Sciences, Division of Neuro-Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
47
|
Robinson AJ, Jain A, Sherman HG, Hague RJM, Rahman R, Sanjuan‐Alberte P, Rawson FJ. Toward Hijacking Bioelectricity in Cancer to Develop New Bioelectronic Medicine. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Andie J. Robinson
- Regenerative Medicine and Cellular Therapies, School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| | - Akhil Jain
- Regenerative Medicine and Cellular Therapies, School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| | - Harry G. Sherman
- Regenerative Medicine and Cellular Therapies, School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| | - Richard J. M. Hague
- Centre for Additive Manufacturing, Faculty of Engineering University of Nottingham Nottingham NG8 1BB UK
| | - Ruman Rahman
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine University of Nottingham Nottingham NG7 2RD UK
| | - Paola Sanjuan‐Alberte
- Regenerative Medicine and Cellular Therapies, School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
- Department of Bioengineering and iBB‐Institute for Bioengineering and Biosciences, Instituto Superior Técnico Universidade de Lisboa Lisbon 1049‐001 Portugal
| | - Frankie J. Rawson
- Regenerative Medicine and Cellular Therapies, School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| |
Collapse
|
48
|
Shawki MM, Elabd S. Tumor treating fields (TTFs) using uninsulated electrodes induce cell death in human non-small cell lung carcinoma (NSCLC) cells. AIMS BIOPHYSICS 2021. [DOI: 10.3934/biophy.2021011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
Saito A, Wada K, Suzuki Y, Nakasono S. The response of the neuronal activity in the somatosensory cortex after high-intensity intermediate-frequency magnetic field exposure to the spinal cord in rats under anesthesia and waking states. Brain Res 2020; 1747:147063. [PMID: 32818531 DOI: 10.1016/j.brainres.2020.147063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/08/2020] [Accepted: 08/14/2020] [Indexed: 12/01/2022]
Abstract
Novel technologies using the intermediate-frequency magnetic field (IF-MF) in living environments are becoming popular with the advance in electricity utilization. However, the biological effects induced by the high-intensity and burst-type IF-MF exposure used in the wireless power transfer technologies for electric vehicles or medical devices, such as the magnetic stimulation techniques, are not well understood. Here, we developed an experimental platform using rats, that combined an 18 kHz, high-intensity (Max. 88 mT), Gaussian-shaped burst IF-MF exposure system with an in vivo extracellular recording system. In this paper, we aimed to report the qualitative differences in stimulus responses in the regions of the somatosensory cortex and peripheral nerve fibers that were induced by the IF-MF exposure to the rat spinal cord. We also report the modulation of the stimulus responses in the somatosensory cortex under anesthesia or waking states. Using this experimental platform, we succeeded in the detection of the motor evoked potentials or the neuronal activity in the somatosensory cortex that was induced by the IF-MF exposure to the spinal cord in rats. Compared to the state of anesthesia, the neuronal activities in the somatosensory cortex was enhanced during the waking state. On the other hand, these neuronal responses could not be confirmed by the IF-MF exposure-related coil sound only. Our experimental results indicated the basic knowledge of the biological responses and excitation mechanisms of the spinal cord stimulation by the IF-MF exposure.
Collapse
Affiliation(s)
- Atsushi Saito
- Biological Environment Sector, Environmental Science Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), 1646 Abiko, Abiko-shi, Chiba, Japan.
| | - Keiji Wada
- Department of Electrical Engineering and Computer Science, Graduate School of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo, Japan.
| | - Yukihisa Suzuki
- Department of Electrical Engineering and Computer Science, Graduate School of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo, Japan.
| | - Satoshi Nakasono
- Biological Environment Sector, Environmental Science Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), 1646 Abiko, Abiko-shi, Chiba, Japan.
| |
Collapse
|
50
|
Carrieri FA, Smack C, Siddiqui I, Kleinberg LR, Tran PT. Tumor Treating Fields: At the Crossroads Between Physics and Biology for Cancer Treatment. Front Oncol 2020; 10:575992. [PMID: 33215030 PMCID: PMC7664989 DOI: 10.3389/fonc.2020.575992] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
Despite extraordinary advances that have been achieved in the last few decades, cancer continues to represent a leading cause of mortality worldwide. Lethal cancer types ultimately become refractory to standard of care approaches; thus, novel effective treatment options are desperately needed. Tumor Treating Fields (TTFields) are an innovative non-invasive regional anti-mitotic treatment modality with minimal systemic toxicity. TTFields are low intensity (1-3 V/cm), intermediate frequency (100-300 kHz) alternating electric fields delivered to cancer cells. In patients, TTFields are applied using FDA-approved transducer arrays, orthogonally positioned on the area surrounding the tumor region, with side effects mostly limited to the skin. The precise molecular mechanism of the anti-tumor effects of TTFields is not well-understood, but preclinical research on TTFields suggests it may act during two phases of mitosis: at metaphase, by disrupting the formation of the mitotic spindle, and at cytokinesis, by dielectrophoretic dislocation of intracellular organelles leading to cell death. This review describes the mechanism of action of TTFields and provides an overview of the most important in vitro studies that investigate the disruptive effects of TTFields in different cancer cells, focusing mainly on anti-mitotic roles. Lastly, we summarize completed and ongoing TTFields clinical trials on a variety of solid tumors.
Collapse
Affiliation(s)
- Francesca A. Carrieri
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caleb Smack
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ismaeel Siddiqui
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Phuoc T. Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Program in Cancer Invasion and Metastasis, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|