1
|
Heo J, Jo Y, Yoon M. Synergistic effects of combined hyperthermia and electric fields treatment in non-small cell lung-cancer (NSCLC) cell lines. Clin Transl Oncol 2025; 27:2014-2023. [PMID: 39436621 DOI: 10.1007/s12094-024-03760-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024]
Abstract
PURPOSE Lung cancer remains a leading cause of cancer-related mortality, with non-small cell lung cancer (NSCLC) being particularly challenging due to poor survival rates, emphasizing the need for new treatments. This study examined the therapeutic effects of combining hyperthermia (HT) with tumor-treating electric fields (TTF) in NSCLC. METHODS Cells were exposed to four different conditions: hyperthermia at 42 °C for 30 min, electric fields at 150 kHz and 0.8 V/cm for 24 h, a combination of both treatments, or no treatment (control). Cell proliferation was measured using WST and colony-formation assays, while apoptosis, DNA damage, and repair protein levels were analyzed via Western blotting. Metastatic potential was evaluated with a transwell assay, and cell migration was assessed using the wound-healing assay. RESULTS The combination therapy significantly inhibited colony formation and reduced cell migration and invasion more effectively than individual treatments. The combined treatment also enhanced apoptosis, as indicated by increased cleaved-PARP and Annexin V levels. In addition, the DNA-damage marker γ-H2AX was elevated, while BRCA1, a protein involved in DNA repair, was significantly downregulated compared to the individual treatments. CONCLUSIONS These results suggest that the enhanced anticancer effects of HT and TTF are due to increased DNA damage and suppression of DNA-repair mechanisms, highlighting the potential of this combination therapy for NSCLC treatment.
Collapse
Affiliation(s)
- Jinju Heo
- Department of Bio-Medical Engineering, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Yunhui Jo
- Institute of Global Health Technology (IGHT), Korea University, Seoul, Republic of Korea
| | - Myonggeun Yoon
- Department of Bio-Medical Engineering, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea.
- FieldCure Ltd., Seoul, 02852, Republic of Korea.
| |
Collapse
|
2
|
Bystrov DA, Volegova DD, Korsakova SA, Salmina AB, Yurchenko SO. Electric Field-Induced Effects in Eukaryotic Cells: Current Progress and Limitations. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40279199 DOI: 10.1089/ten.teb.2025.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Electric fields (EFs) offer a powerful tool for manipulating cells and modulating their behavior, holding significant promise for regenerative medicine and cell biology. We provide a comprehensive overview of the effects of different types of EF on eukaryotic cells with the special focus on physical mechanisms and signaling pathways involved. Direct current EF induces electrophoresis and electroosmosis, influencing cell migration, proliferation, and differentiation. Alternating current EF, through dielectric polarization and dielectrophoresis, enables cell manipulation, trapping, and sorting. Pulsed EF, particularly high-intensity, short-duration pulses, induces reversible and irreversible electroporation, facilitating drug and gene delivery. The review covers some technological aspects of EF generation, emphasizing the importance of experimental setups, and integration with microfluidic platforms for high-throughput analysis and precise manipulations. Furthermore, the synergistic potential of combining EFs with optical tweezers is highlighted, enabling fine-tuned control of cell positioning, intercellular interactions, and measurement of biophysical properties. Finally, the review addresses limitations of EF application, such as field heterogeneity and potential side effects, and outlines the directions for future studies, including developing the minimally invasive delivery methods.
Collapse
Affiliation(s)
- Daniil A Bystrov
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| | - Daria D Volegova
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| | - Sofia A Korsakova
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| | - Alla B Salmina
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
| | - Stanislav O Yurchenko
- Center "Soft Matter and Physics of Fluids," Bauman Moscow State Technical University, Moscow, Russia
| |
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
|
Heo J, Jo Y, Yoon M. Enhanced anti-tumor effects of combined electric fields, cabozantinib, and radiation therapy in metastatic renal cell carcinoma. Clin Transl Oncol 2025:10.1007/s12094-025-03898-x. [PMID: 40126769 DOI: 10.1007/s12094-025-03898-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: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
PURPOSE This study aimed to evaluate the therapeutic potential of combining cabozantinib, electric fields (EFs; also called Tumor Treating Fields [TTFields]), and radiation in the treatment of metastatic renal cell carcinoma (RCC), focusing on overcoming resistance to conventional monotherapeutic regimens. METHODS Human renal cancer cell lines (A498, Caki-1) were treated with cabozantinib (10 µM) for at least 6 h, TTFields (200 kHz, 0.8 V/cm) for 24 h, and radiation (3 Gy), both individually and in combination. Cellular responses, including proliferation, apoptosis, and metastatic potential, were analyzed by flow cytometry and Transwell assays. RESULTS The combination of cabozantinib, TTFields, and radiation exhibited synergistic effects, significantly reducing cell proliferation, enhancing apoptosis, and suppressing metastatic capacity compared with individual treatments. Triple therapy resulted in marked inhibition of metastasis-related markers and changes in apoptotic profiles compared with the control group. CONCLUSION This multimodal therapy demonstrated superior efficacy in reducing the metastatic potential and prolonging the survival of RCC cells, offering a promising approach to overcoming treatment resistance in patients with metastatic RCC.
Collapse
Affiliation(s)
- Jinju Heo
- Department of Bio-Medical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Yunhui Jo
- Institute of Global Health Technology (IGHT), Korea University, Seoul, Republic of Korea
| | - Myonggeun Yoon
- Department of Bio-Medical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea.
- FieldCure Ltd, Seoul, 02852, Republic of Korea.
| |
Collapse
|
5
|
Lin W, Wang Y, Li M, Feng J, Yue Y, Yu J, Hu Y, Suo Y. Tumor treating fields enhance anti-PD therapy by improving CCL2/8 and CXCL9/CXCL10 expression through inducing immunogenic cell death in NSCLC models. BMC Cancer 2025; 25:489. [PMID: 40098106 PMCID: PMC11912744 DOI: 10.1186/s12885-025-13859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. Tumor treating fields (TTFields) combined with anti-PD immunotherapy offers a promising strategy to address this issue. Nevertheless, the mechanism of action (MOA) of TTFields therapy combined with anti-PD immunotherapy in NSCLC has not been thoroughly investigated. This study aims to elucidate the MOA of the combined therapy from the aspect of improving the tumor immune microenvironment (TIME). METHODS Using a mouse model of NSCLC, we tested the efficacy of TTFields therapy with anti-PD-1 and anti-PD-L1 immunotherapy. By RNA-seq, the differential genes and signaling pathways between combination therapy and anti-PD therapy groups were studied. In-vitro experiments validated the effects of TTFields on tumor cells for CD4+ T cell and CD8+ T cell infiltration, as well as the expression of tumor immunogenic death related genes and chemokines. RESULTS Combining TTFields with anti-PD-1 reduced tumor weight and volume, respectively, compared to controls (p < 0.05). RNA-seq analysis revealed 1,745 differentially expressed genes (DEGs) in the combination therapy group versus controls, including upregulated immune pathways and immunogenic cell death (ICD) associated genes. Further study showed that the combination therapy resulted in increased T cell infiltration compared to anti-PD immunotherapy alone, and TTFields induced higher level expression of ATP, HMGB1, CCL2, CCL8, CXCL9, and CXCL10 and inflammatory cytokines than control group. These effects collectively contributed to the altered TIME, and finally potentiated the efficacy of anti-PD therapy. CONCLUSIONS TTFields enhance the effectiveness of anti-PD immunotherapy by improving CD4+ T cells and CD8+ T infiltration via inducing ICD to increase CCL2/8 and CXCL9/CXCL10 expression of tumor cells. This study provides theoretical basis and new insights for evaluating the effectiveness of TTFields combined with anti-PD therapy for NSCLC.
Collapse
Affiliation(s)
- Wei Lin
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China.
- School of Clinical and Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Medicine and Health Key Laboratory of Rheumatism, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China.
- Department of Critical-care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| | - Yingying Wang
- Healthy Life Innovation Medical Technology Co., Ltd, Wuxi, 214174, China
| | - Minghao Li
- School of Clinical and Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jingjing Feng
- Healthy Life Innovation Medical Technology Co., Ltd, Wuxi, 214174, China
| | - Ying Yue
- Healthy Life Innovation Medical Technology Co., Ltd, Wuxi, 214174, China
| | - Jing Yu
- Healthy Life Innovation Medical Technology Co., Ltd, Wuxi, 214174, China
| | - Yanjiang Hu
- Department of Thoracic Surgery, Liyang People's Hospital, Liyang, 213300, China.
| | - Yuanzhen Suo
- Healthy Life Innovation Medical Technology Co., Ltd, Wuxi, 214174, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
6
|
Qi Y, Cao J, Jiang M, Lin Y, Li W, Li B. HSP27/IL-6 axis promotes OSCC chemoresistance, invasion and migration by orchestrating macrophages via a positive feedback loop. Cell Biol Toxicol 2025; 41:36. [PMID: 39873845 PMCID: PMC11775009 DOI: 10.1007/s10565-024-09983-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/21/2024] [Indexed: 01/30/2025]
Abstract
Novel strategies to disrupt tumor progression have emerged from studying the interactions between tumor cells and tumor-associated macrophages (TAMs). However, the molecular mechanisms of interactions between tumor cells and TAMs underlying oral squamous cell carcinoma (OSCC) progression have not been fully elucidated. This study explored the molecular mechanism of the HSP27/IL-6 axis in OSCC chemoresistance, invasion, and migration. Here, we demonstrated the higher expression of HSP27 in OSCC cells. Paracrine HSP27 from OSCC cells enhanced chemoresistance, invasion, migration, and EMT in OSCC by inducing M2 polarization and IL-6 secretion in TAMs. HSP27 and IL-6 established a positive feedback loop between OSCC cells and M2 TAMs. TAMs-derived IL-6 orchestrated OSCC stemness and chemoresistance through upregulating β-catenin and CD44, and enhanced OSCC invasion, migration, and EMT via autocrine HSP27/TLR4 signaling. Collectively, HSP27/IL-6 axis facilitates OSCC chemoresistance, invasion, and migration by orchestrating macrophages through a positive feedback loop. We identify the regulatory mechanism underlying the interaction and crosstalk between OSCC cells and TAMs mediated by the HSP27/IL-6 axis. Targeting the HSP27/IL-6 axis could be a promising treatment strategy for OSCC patients, potentially controlling disease progression and improving prognosis and recurrence outcomes.
Collapse
Affiliation(s)
- Ying Qi
- Department of Oral Anatomy and Physiology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Jilin University, Changchun, 130021, China
| | - Juan Cao
- Department of Oral Anatomy and Physiology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Jilin University, Changchun, 130021, China
| | - Mingjing Jiang
- Department of Oral Anatomy and Physiology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Jilin University, Changchun, 130021, China
| | - Ying Lin
- Department of Oral Anatomy and Physiology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Jilin University, Changchun, 130021, China
| | - Weibo Li
- Department of Oral Anatomy and Physiology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Jilin University, Changchun, 130021, China
| | - Bo Li
- Department of Oral Anatomy and Physiology, Hospital of Stomatology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Jilin University, Changchun, 130021, China.
| |
Collapse
|
7
|
Amodeo R, Morosi L, Meroni M, Bello E, Timo S, Frapolli R, D’Incalci M, Lupi M. Tumor Treating Fields enhance chemotherapy efficacy by increasing cellular drug uptake and retention in mesothelioma cells. Am J Cancer Res 2025; 15:271-285. [PMID: 39949944 PMCID: PMC11815374 DOI: 10.62347/odwl5634] [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: 07/05/2024] [Accepted: 09/04/2024] [Indexed: 02/16/2025] Open
Abstract
Tumor Treating Fields (TTFields) applied with standard chemotherapy have been approved for the first-line treatment of unresectable pleural mesothelioma (PM), an aggressive malignancy with limited effective therapy options. In this study, we demonstrated that the simultaneous exposure to TTFields and doxorubicin or vinorelbine enhanced treatment efficacy in patient-derived PM cells by increasing intracellular drug concentrations. This was achieved by modulating several genes that encode transport proteins, such as the downregulation of P-glycoprotein (P-gp). Using specific, sensitive and quantitative analytical techniques, we observed a more than 70% increase in intracellular concentrations of doxorubicin and vinorelbine in samples treated with TTFields, and a greater than 50% increase in drug uptake in cells exposed to TTFields and pemetrexed. This result indicates that the increased drug concentration observed in TTFields treated cells is significant not only for drugs that are P-gp substrates but also suggests that TTFields could potentially affect other efflux pumps. However, the co-exposure to the drug and TTFields was critical to increasing intracellular drug levels, highlighting the necessity of concurrent use with drugs to enhance the antiproliferative effects of treatment. The in vitro findings were further corroborated by in vivo pharmacokinetic experiments in mice subcutaneously injected with epithelioid PM tumors. Indeed, a 30% increase in intratumor concentrations was observed when vinorelbine was administered with TTFields. Our findings suggest that TTFields could be a well-tolerated approach for enhancing intratumoral drug levels and potentially achieving a more significant therapeutic impact on PM treatment.
Collapse
Affiliation(s)
- Rosy Amodeo
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research HospitalRozzano, Milano, Italy
- Department of Biomedical Sciences, Humanitas UniversityPieve Emanuele, Milano, Italy
| | - Lavinia Morosi
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research HospitalRozzano, Milano, Italy
| | - Marina Meroni
- Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCSMilano, Italy
| | - Ezia Bello
- Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCSMilano, Italy
| | - Sara Timo
- Metabolomics and Pharmacokinetics Unit, IRCCS Humanitas Research HospitalRozzano, Milano, Italy
| | - Roberta Frapolli
- Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCSMilano, Italy
| | - Maurizio D’Incalci
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research HospitalRozzano, Milano, Italy
- Department of Biomedical Sciences, Humanitas UniversityPieve Emanuele, Milano, Italy
| | - Monica Lupi
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research HospitalRozzano, Milano, Italy
| |
Collapse
|
8
|
Tanzhu G, Chen L, Ning J, Xue W, Wang C, Xiao G, Yang J, Zhou R. Metastatic brain tumors: from development to cutting-edge treatment. MedComm (Beijing) 2025; 6:e70020. [PMID: 39712454 PMCID: PMC11661909 DOI: 10.1002/mco2.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 12/24/2024] Open
Abstract
Metastatic brain tumors, also called brain metastasis (BM), represent a challenging complication of advanced tumors. Tumors that commonly metastasize to the brain include lung cancer and breast cancer. In recent years, the prognosis for BM patients has improved, and significant advancements have been made in both clinical and preclinical research. This review focuses on BM originating from lung cancer and breast cancer. We briefly overview the history and epidemiology of BM, as well as the current diagnostic and treatment paradigms. Additionally, we summarize multiomics evidence on the mechanisms of tumor occurrence and development in the era of artificial intelligence and discuss the role of the tumor microenvironment. Preclinically, we introduce the establishment of BM models, detailed molecular mechanisms, and cutting-edge treatment methods. BM is primarily treated with a comprehensive approach, including local treatments such as surgery and radiotherapy. For lung cancer, targeted therapy and immunotherapy have shown efficacy, while in breast cancer, monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates are effective in BM. Multiomics approaches assist in clinical diagnosis and treatment, revealing the complex mechanisms of BM. Moreover, preclinical agents often need to cross the blood-brain barrier to achieve high intracranial concentrations, including small-molecule inhibitors, nanoparticles, and peptide drugs. Addressing BM is imperative.
Collapse
Affiliation(s)
- Guilong Tanzhu
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Liu Chen
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jiaoyang Ning
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Wenxiang Xue
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunJilinChina
| | - Ce Wang
- Department of RadiologyChina‐Japan Friendship HospitalBeijingChina
| | - Gang Xiao
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
| | - Jie Yang
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Department of DermatologyXiangya HospitalCentral South UniversityChangshaChina
| | - Rongrong Zhou
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- Xiangya Lung Cancer CenterXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan ProvinceChina
| |
Collapse
|
9
|
Costa FP, Wiedenmann B, Schöll E, Tuszynski J. Emerging cancer therapies: targeting physiological networks and cellular bioelectrical differences with non-thermal systemic electromagnetic fields in the human body - a comprehensive review. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1483401. [PMID: 39720338 PMCID: PMC11666389 DOI: 10.3389/fnetp.2024.1483401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024]
Abstract
A steadily increasing number of publications support the concept of physiological networks, and how cellular bioelectrical properties drive cell proliferation and cell synchronization. All cells, especially cancer cells, are known to possess characteristic electrical properties critical for physiological behavior, with major differences between normal and cancer cell counterparts. This opportunity can be explored as a novel treatment modality in Oncology. Cancer cells exhibit autonomous oscillations, deviating from normal rhythms. In this context, a shift from a static view of cellular processes is required for a better understanding of the dynamic connections between cellular metabolism, gene expression, cell signaling and membrane polarization as states in constant flux in realistic human models. In oncology, radiofrequency electromagnetic fields have produced sustained responses and improved quality of life in cancer patients with minimal side effects. This review aims to show how non-thermal systemic radiofrequency electromagnetic fields leads to promising therapeutic responses at cellular and tissue levels in humans, supporting this newly emerging cancer treatment modality with early favorable clinical experience specifically in advanced cancer.
Collapse
Affiliation(s)
| | | | - Eckehard Schöll
- Institut für Theoretische Physik, Technische Universität Berlin, Berlin, Germany
| | - Jack Tuszynski
- Department of Physics, University of Alberta, Edmonton, AB, Canada
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Turin, Italy
- Department of Data Science and Engineering, The Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
10
|
Djamgoz MBA. Electrical excitability of cancer cells-CELEX model updated. Cancer Metastasis Rev 2024; 43:1579-1591. [PMID: 38976181 PMCID: PMC11554705 DOI: 10.1007/s10555-024-10195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
The normal functioning of every cell in the body depends on its bioelectric properties and many diseases are caused by genetic and/or epigenetic dysregulation of the underlying ion channels. Metastasis, the main cause of death from cancer, is a complex multi-stage process in which cells break away from a primary tumour, invade the surrounding tissues, enter the circulation by encountering a blood vessel and spread around the body, ultimately lodging in distant organs and reproliferating to form secondary tumours leading to devastating organ failure. Such cellular behaviours are well known to involve ion channels. The CELEX model offers a novel insight to metastasis where it is the electrical excitation of the cancer cells that is responsible for their aggressive and invasive behaviour. In turn, the hyperexcitability is underpinned by concomitant upregulation of functional voltage-gated sodium channels and downregulation of voltage-gated potassium channels. Here, we update the in vitro and in vivo evidence in favour of the CELEX model for carcinomas. The results are unequivocal for the sodium channel. The potassium channel arm is also broadly supported by existing evidence although these data are complicated by the impact of the channels on the membrane potential and consequent secondary effects. Finally, consistent with the CELEX model, we show (i) that carcinomas are indeed electrically excitable and capable of generating action potentials and (ii) that combination of a sodium channel inhibitor and a potassium channel opener can produce a strong, additive anti-invasive effect. We discuss the possible clinical implications of the CELEX model in managing cancer.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
11
|
Wen J, Xiong L, Wang S, Qiu X, Cui J, Peng F, Liu X, Lu J, Bian H, Chen D, Chang J, Yao Z, Fan S, Zhou D, Li Z, Liu J, Liu H, Chen X, Chen L. Prediction of intracranial electric field strength and analysis of treatment protocols in tumor electric field therapy targeting gliomas of the brain. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 258:108490. [PMID: 39520874 DOI: 10.1016/j.cmpb.2024.108490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND OBJECTIVE Tumor Electric Field Therapy (TEFT) is a new treatment for glioblastoma cells with significant effect and few side effects. However, it is difficult to directly measure the intracranial electric field generated by TEFT, and the inability to control the electric field intensity distribution in the tumor target area also limits the clinical therapeutic effect of TEFT. It is a safe and effective way to construct an efficient and accurate prediction model of intracranial electric field intensity of TEFT by numerical simulation. METHODS Different from the traditional methods, in this study, the brain tissue was segmented based on the MRI data of patients with retained spatial location information, and the spatial position of the brain tissue was given the corresponding electrical parameters after segmentation. Then, a single geometric model of the head profile with the transducer array is constructed, which is assembled with an electrical parameter matrix containing tissue position information. After applying boundary conditions on the transducer, the intracranial electric field intensity could be solved in the frequency domain. The effects of transducer array mode, load voltage and voltage frequency on the intracranial electric field strength were further analyzed. Finally, planning system software was developed for optimizing TEFT treatment regimens for patients. RESULTS Experimental validation and comparison with existing results demonstrate the proposed method has a more efficient and pervasive modeling approach with higher computational accuracy while preserving the details of MRI brain tissue structure completely. In the optimization analysis of treatment protocols, it was found that increasing the load voltage could effectively increase the electric field intensity in the target area, while the effect of voltage frequency on the electric field intensity was very limited. CONCLUSIONS The results showed that adjusting the transducer array mode was the key method for making targeted treatment plans. The proposed method is capable prediction of intracranial electric field strength with high accuracy and provide guidance for the design of the TEFT therapy process. This study provides a valuable reference for the application of TEFT in clinical practice.
Collapse
Affiliation(s)
- Jun Wen
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Lingzhi Xiong
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Shulu Wang
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Xiaoguang Qiu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Jianqiao Cui
- Hunan Drug Inspection Center, Changsha, Hunan, China
| | - Fan Peng
- Public Course Teaching Department, Changsha Health Vocational College, Changsha 410100, China
| | - Xiang Liu
- Hunan Drug Inspection Center, Changsha, Hunan, China
| | - Jian Lu
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Haikuo Bian
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Jiusheng Chang
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Zhengxi Yao
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Sheng Fan
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Dan Zhou
- Hunan An Tai Kang Cheng Biotechnology Co., Changsha, Hunan, China
| | - Ze Li
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Jialin Liu
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Hongyu Liu
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Xu Chen
- The First Clinical College, China Medical University, Shenyang, China
| | - Ling Chen
- Department of Neurosurgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
12
|
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
|
13
|
Peng J, Han Y, Wang L. Opening neural gateways: old dog now has new tricks. Front Pharmacol 2024; 15:1389383. [PMID: 39011502 PMCID: PMC11246957 DOI: 10.3389/fphar.2024.1389383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Affiliation(s)
- Jiamin Peng
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yong Han
- Department of Thoracic Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Ligang Wang
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Wu H, Zhou F, Gao W, Chen P, Wei Y, Wang F, Zhao H. Current status and research progress of minimally invasive treatment of glioma. Front Oncol 2024; 14:1383958. [PMID: 38835394 PMCID: PMC11148461 DOI: 10.3389/fonc.2024.1383958] [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: 02/08/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Glioma has a high malignant degree and poor prognosis, which seriously affects the prognosis of patients. Traditional treatment methods mainly include craniotomy tumor resection, postoperative radiotherapy and chemotherapy. Although above methods have achieved remarkable curative effect, they still have certain limitations and adverse reactions. With the introduction of the concept of minimally invasive surgery and its clinical application as well as the development and progress of imaging technology, minimally invasive treatment of glioma has become a research hotspot in the field of neuromedicine, including photothermal treatment, photodynamic therapy, laser-induced thermal theraphy and TT-Fields of tumor. These therapeutic methods possess the advantages of precision, minimally invasive, quick recovery and significant curative effect, and have been widely used in clinical practice. The purpose of this review is to introduce the progress of minimally invasive treatment of glioma in recent years and the achievements and prospects for the future.
Collapse
Affiliation(s)
- Hao Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Feng Zhou
- Department of Neurosurgery, The First Hospital of Yu Lin, Yulin, China
| | - Wenwen Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Peng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Yao Wei
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Fenglu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, China
| | - Haikang Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, China
| |
Collapse
|
15
|
Lin H, Liu C, Hu A, Zhang D, Yang H, Mao Y. Understanding the immunosuppressive microenvironment of glioma: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:31. [PMID: 38720342 PMCID: PMC11077829 DOI: 10.1186/s13045-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4-8%) of CD45+ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Collapse
Affiliation(s)
- Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
16
|
Xing Y, Yasinjan F, Cui J, Peng Y, He M, Liu W, Hong X. Advancements and current trends in tumor treating fields: a scientometric analysis. Int J Surg 2024; 110:2978-2991. [PMID: 38349201 PMCID: PMC11093503 DOI: 10.1097/js9.0000000000001151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/26/2024] [Indexed: 05/16/2024]
Abstract
Tumor treating fields (TTFields) therapy is a novel and effective noninvasive cancer therapy, and it has been approved by FDA in the treatment of recurrent and newly diagnosed glioblastoma, and malignant pleural mesothelioma. Moreover, TTFields therapy has been widely studied in both clinical trials and preclinical studies in recent years. Based on its high efficacy, research on TTFields therapy has been a hot topic. Thus, the authors made this scientometric analysis of TTfields to reveal the scientometric distributions such as annual publications and citations, countries and institutions, authors, journals, references, and more importantly, research status and hot topics of the field. In recent years, publication numbers have been stable at high values, and citation numbers have been increasing greatly. The United States and Israel were the top two countries with the highest publication numbers, followed by Germany and Switzerland. Scientometric analyses of keywords indicated that clinical applications and antitumor mechanisms are probably the two main parts of current research on TTfields. Most clinical trials of TTfields focus on the treatment of glioblastoma. And a variety of other cancers such as lung cancer especially nonsmall cell lung cancer, hepatic cancer, other brain tumors, etc. have also been studied in both clinical trials and preclinical studies.
Collapse
Affiliation(s)
- Yang Xing
- Department of Neurosurgery, The First Hospital of Jilin University
| | - Feroza Yasinjan
- Department of Neurosurgery, The First Hospital of Jilin University
| | - Jiayue Cui
- Department of Histology and Embryology, College of Basic Medical Sciences
| | - Yizhao Peng
- Department of Neurosurgery, The First Hospital of Jilin University
| | - Minghua He
- College of Computer Science and Technology, Jilin University, Changchun, People’s Republic of China
| | - Wenhui Liu
- Department of Histology and Embryology, College of Basic Medical Sciences
| | - Xinyu Hong
- Department of Neurosurgery, The First Hospital of Jilin University
| |
Collapse
|
17
|
Lan J, Liu Y, Chen J, Liu H, Feng Y, Liu J, Chen L. Advanced tumor electric fields therapy: A review of innovative research and development and prospect of application in glioblastoma. CNS Neurosci Ther 2024; 30:e14720. [PMID: 38715344 PMCID: PMC11077002 DOI: 10.1111/cns.14720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive malignant tumor with a high mortality rate and is the most prevalent primary intracranial tumor that remains incurable. The current standard treatment, which involves surgery along with concurrent radiotherapy and chemotherapy, only yields a survival time of 14-16 months. However, the introduction of tumor electric fields therapy (TEFT) has provided a glimmer of hope for patients with newly diagnosed and recurrent GBM, as it has been shown to extend the median survival time to 20 months. The combination of TEFT and other advanced therapies is a promising trend in the field of GBM, facilitated by advancements in medical technology. AIMS In this review, we provide a concise overview of the mechanism and efficacy of TEFT. In addition, we mainly discussed the innovation of TEFT and our proposed blueprint for TEFT implementation. CONCLUSION Tumor electric fields therapy is an effective and highly promising treatment modality for GBM. The full therapeutic potential of TEFT can be exploited by combined with other innovative technologies and treatments.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| | - Yuyang Liu
- Medical School of Chinese PLABeijingChina
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Junyi Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Hongyu Liu
- Medical School of Chinese PLABeijingChina
- Department of NeurosurgeryHainan Hospital of Chinese PLA General HospitalHainanChina
| | - Yaping Feng
- Department of Neurosurgery920th Hospital of Joint Logistics Support ForceKunmingChina
| | - Jialin Liu
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Ling Chen
- Department of NeurosurgeryChinese PLA General HospitalBeijingChina
- School of MedicineNankai UniversityTianjinChina
- Medical School of Chinese PLABeijingChina
| |
Collapse
|
18
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
19
|
Xiang XW, Liu HT, Tao XN, Zeng YL, Liu J, Wang C, Yu SX, Zhao H, Liu YJ, Liu KF. Glioblastoma behavior study under different frequency electromagnetic field. iScience 2023; 26:108575. [PMID: 38125027 PMCID: PMC10730381 DOI: 10.1016/j.isci.2023.108575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/06/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
The tumor-treating fields (TTFields) technology has revolutionized the management of recurrent and newly diagnosed glioblastoma (GBM) cases. To ameliorate this treatment modality for GBM and other oncological conditions, it is necessary to understand the biophysical principles of TTFields better. In this study, we further analyzed the mechanism of the electromagnetic exposure with varying frequencies and electric field strengths on cells in mitosis, specifically in telophase. In reference to previous studies, an intuitive finite element model of the mitotic cell was built for electromagnetic simulations, predicting a local increase in the cleavage furrow region, which may help explain TTFields' anti-proliferative effects. Cell experiments confirmed that the reduction in proliferation and migration of glioma cell by TTFields was in a frequency- and field-strength-dependent manner. This work provides unique insights into the selection of frequencies in the anti-proliferative effect of TTFields on tumors, which could improve the application of TTFields.
Collapse
Affiliation(s)
- Xiao-Wei Xiang
- Academy for engineering & technology, Fudan University, Shanghai 200433, China
| | - Hao-Tian Liu
- Academy for engineering & technology, Fudan University, Shanghai 200433, China
| | - Xiao-Nan Tao
- School of information science and technology, Fudan University, Shanghai 200433, China
| | - Yu-Lian Zeng
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Jing Liu
- School of information science and technology, Fudan University, Shanghai 200433, China
| | - Chen Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hui Zhao
- School of information science and technology, Fudan University, Shanghai 200433, China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ke-Fu Liu
- School of information science and technology, Fudan University, Shanghai 200433, China
| |
Collapse
|
20
|
Szklener K, Bilski M, Nieoczym K, Mańdziuk D, Mańdziuk S. Enhancing glioblastoma treatment through the integration of tumor-treating fields. Front Oncol 2023; 13:1274587. [PMID: 37916157 PMCID: PMC10616854 DOI: 10.3389/fonc.2023.1274587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/28/2023] [Indexed: 11/03/2023] Open
Abstract
Glioblastoma (GBM) represents a significant therapeutic challenge due to its aggressive nature. Tumor Treating Fields (TTFields) present a promising approach to GBM therapy. The primary mechanism of TTFields, an antimitotic effect, alongside numerous indirect effects including increased cell membrane permeability, signifies their potential in combination with other treatment modalities. Current combinations often include chemotherapy, particularly with temozolomide (TMZ), however, emerging data suggests potential synergy with targeted therapies, radiotherapy, and immunotherapy as well. TTFields display minimal side effects, predominantly skin-related, posing no significant barrier to combined therapies. The effectiveness of TTFields in GBM treatment has been demonstrated through several post-registration studies, advocating for continued research to optimize overall survival (OS) and progression-free survival (PFS) in patients, as opposed to focusing solely on quality of life.
Collapse
Affiliation(s)
- Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Mateusz Bilski
- Department of Radiotherapy, Medical University of Lublin, Lublin, Poland
| | - Karolina Nieoczym
- Student Scientific Association at the Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Dominika Mańdziuk
- Student Scientific Association at the Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Sławomir Mańdziuk
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
21
|
Chen S, Cao R, Xiang L, Li Z, Chen H, Zhang J, Feng X. Research progress in nucleus-targeted tumor therapy. Biomater Sci 2023; 11:6436-6456. [PMID: 37609783 DOI: 10.1039/d3bm01116j] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The nucleus is considered the most important organelle in the cell as it plays a central role in controlling cell reproduction, metabolism, and the cell cycle. The successful delivery of drugs into the nucleus can achieve excellent therapeutic effects, which reveals the potential of nucleus-targeted therapy in precision medicine. However, the transportation of therapeutics into the nucleus remains a significant challenge due to various biological barriers. Herein, we summarize the recent progress in the nucleus-targeted drug delivery system (NDDS). The structures of the nucleus and nuclear envelope are first described in order to understand the mechanisms by which drugs cross the nuclear envelope. Then, various drug delivery strategies based on the mechanisms and their applications are discussed. Finally, the challenges and solutions in the field of nucleus-targeted drug delivery are raised for developing a more efficient NDDS and promoting its clinical transformation.
Collapse
Affiliation(s)
- Shaofeng Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Rumeng Cao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ling Xiang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ziyi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Hui Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Jiumeng Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Xuli Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| |
Collapse
|
22
|
Li X, Liu K, Xing L, Rubinsky B. A review of tumor treating fields (TTFields): advancements in clinical applications and mechanistic insights. Radiol Oncol 2023; 57:279-291. [PMID: 37665740 PMCID: PMC10476910 DOI: 10.2478/raon-2023-0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/04/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Tumor Treating Fields (TTFields) is a non-invasive modality for cancer treatment that utilizes a specific sinusoidal electric field ranging from 100 kHz to 300 kHz, with an intensity of 1 V/cm to 3 V/cm. Its purpose is to inhibit cancer cell proliferation and induce cell death. Despite promising outcomes from clinical trials, TTFields have received FDA approval for the treatment of glioblastoma multiforme (GBM) and malignant pleural mesothelioma (MPM). Nevertheless, global acceptance of TTFields remains limited. To enhance its clinical application in other types of cancer and gain a better understanding of its mechanisms of action, this review aims to summarize the current research status by examining existing literature on TTFields' clinical trials and mechanism studies. CONCLUSIONS Through this comprehensive review, we seek to stimulate novel ideas and provide physicians, patients, and researchers with a better comprehension of the development of TTFields and its potential applications in cancer treatment.
Collapse
Affiliation(s)
- Xing Li
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nan Jing, Jiang Su, China
| | - Kaida Liu
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nan Jing, Jiang Su, China
| | - Lidong Xing
- College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nan Jing, Jiang Su, China
| | - Boris Rubinsky
- Department of Mechanical Engineering, University of California Berkeley, BerkeleyCA, United States of America
| |
Collapse
|
23
|
Lee WS, Jang Y, Cho A, Kim YB, Bu YH, Yang S, Kim EH. Effectiveness of tumor‑treating fields to reduce the proliferation and migration of liposarcoma cell lines. Exp Ther Med 2023; 26:363. [PMID: 37408858 PMCID: PMC10318604 DOI: 10.3892/etm.2023.12062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/21/2023] [Indexed: 07/07/2023] Open
Abstract
Liposarcoma (LPS) is a rare type of soft tissue sarcoma that constitutes 20% of all sarcoma cases in adults. Effective therapeutic protocols for human LPS are not well-defined. Tumor-treating fields (TTFields) are a novel and upcoming field for antitumor therapy. TTFields combined with chemoradiotherapy have proven to be more effective than TTFields combined with radiotherapy or chemotherapy alone. The present study aimed to assess the effectiveness of TTFields in inhibiting cell proliferation and viability for the anticancer treatment of LPS. The present study used TTFields (frequency, 150 kHz; intensity, 1.0 V/cm) to treat two LPS cell lines (94T778 and SW872) and analyzed the antitumor effects. According to trypan blue and MTT assay results, TTFields markedly reduced the viability and proliferation of LPS cell lines along with the formation of colonies in three-dimensional culture. Based on the Transwell chamber assay, TTFields treatment also markedly reduced the migration of LPS cells. Furthermore, as shown by the higher activation of caspase-3 in the Caspase-3 activity assay and the results of the reactive oxygen species (ROS) assay, TTFields increased the formation of ROS in the cells and enhanced the proportion of apoptotic cells. The present study also investigated the inhibitory effect of TTFields in combination with doxorubicin (DOX) on the migratory capacity of tumor cells. The results demonstrated that TTFields treatment synergistically induced the ROS-induced apoptosis of LPS cancer cell lines and inhibited their migratory behavior. In conclusion, the present study demonstrated the potential of TTFields in improving the sensitivity of LPS cancer cells, which may lay the foundation for future clinical trials of this combination treatment strategy.
Collapse
Affiliation(s)
- Won Seok Lee
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Yoonjung Jang
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Gyeongsangbuk-do 42988, Republic of Korea
| | - Ahyeon Cho
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Yu Bin Kim
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Young Hyun Bu
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Somi Yang
- School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, Gyeongsangbuk-do 42472, Republic of Korea
| |
Collapse
|
24
|
Aurora B Kinase Inhibition by AZD1152 Concomitant with Tumor Treating Fields Is Effective in the Treatment of Cultures from Primary and Recurrent Glioblastomas. Int J Mol Sci 2023; 24:ijms24055016. [PMID: 36902447 PMCID: PMC10003311 DOI: 10.3390/ijms24055016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Tumor Treating Fields (TTFields) were incorporated into the treatment of glioblastoma, the most malignant brain tumor, after showing an effect on progression-free and overall survival in a phase III clinical trial. The combination of TTFields and an antimitotic drug might further improve this approach. Here, we tested the combination of TTFields with AZD1152, an Aurora B kinase inhibitor, in primary cultures of newly diagnosed (ndGBM) and recurrent glioblastoma (rGBM). AZD1152 concentration was titrated for each cell line and 5-30 nM were used alone or in addition to TTFields (1.6 V/cm RMS; 200 kHz) applied for 72 h using the inovitro™ system. Cell morphological changes were visualized by conventional and confocal laser microscopy. The cytotoxic effects were determined by cell viability assays. Primary cultures of ndGBM and rGBM varied in p53 mutational status; ploidy; EGFR expression and MGMT-promoter methylation status. Nevertheless; in all primary cultures; a significant cytotoxic effect was found following TTFields treatment alone and in all but one, a significant effect after treatment with AZD1152 alone was also observed. Moreover, in all primary cultures the combined treatment had the most pronounced cytotoxic effect in parallel with morphological changes. The combined treatment of TTFields and AZD1152 led to a significant reduction in the number of ndGBM and rGBM cells compared to each treatment alone. Further evaluation of this approach, which has to be considered as a proof of concept, is warranted, before entering into early clinical trials.
Collapse
|