151
|
Investigation of the Electrical Properties of Microtubule Ensembles under Cell-Like Conditions. NANOMATERIALS 2020; 10:nano10020265. [PMID: 32033331 PMCID: PMC7075204 DOI: 10.3390/nano10020265] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 01/01/2023]
Abstract
Microtubules are hollow cylindrical polymers composed of the highly negatively-charged (~23e), high dipole moment (1750 D) protein α, β- tubulin. While the roles of microtubules in chromosomal segregation, macromolecular transport, and cell migration are relatively well-understood, studies on the electrical properties of microtubules have only recently gained strong interest. Here, we show that while microtubules at physiological concentrations increase solution capacitance, free tubulin has no appreciable effect. Further, we observed a decrease in electrical resistance of solution, with charge transport peaking between 20-60 Hz in the presence of microtubules, consistent with recent findings that microtubules exhibit electric oscillations at such low frequencies. We were able to quantify the capacitance and resistance of the microtubules (MT) network at physiological tubulin concentrations to be 1.27 × 10-5 F and 9.74 × 104 Ω. Our results show that in addition to macromolecular transport, microtubules also act as charge storage devices through counterionic condensation across a broad frequency spectrum. We conclude with a hypothesis of an electrically tunable cytoskeleton where the dielectric properties of tubulin are polymerisation-state dependent.
Collapse
|
152
|
Affiliation(s)
- M Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
153
|
Jo Y, Sung J, Jeong H, Hong S, Jeong YK, Kim EH, Yoon M. Effectiveness of a Fractionated Therapy Scheme in Tumor Treating Fields Therapy. Technol Cancer Res Treat 2019; 18:1533033819845008. [PMID: 31072204 PMCID: PMC6515848 DOI: 10.1177/1533033819845008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
This study aimed to evaluate the biological effectiveness of cancer therapy with tumor treating fields using a fractionated treatment scheme that was originally designed for radiotherapy. Discontinuous fractional tumor treating fields of an intensity of 0.9 to 1.2 V/cm and a frequency of 150 KHz were applied to U373 cancer cells and IEC6 normal cells for 3 days, with durations of 3, 6, 12, or 24 h/d. As the treatment duration of the tumor treating fields increased from 3 to 24 h/d, the relative tumor cell (U373) number (% of control) reduced in proportion to the treatment duration. Compared to a 25% cell number reduction (75% of control) for the group of 6 h/d treatment at 1.2 V/cm, only 5% (70% of control) and 8% (67% of control) of additional reductions were observed for the group of 12 and 24 h/d treatment, respectively. This experimental result indicates that the dependence on treatment duration in tumor cell inhibition was weakened distinctly at treatment duration over 6 h/d. For normal cells (IEC6), the relative cell number corresponding to the treatment time of the tumor treating fields at 1.2 V/cm of electric field strength was not decreased much for the treatment times of 3, 6, and 12 h/d, revealing 93.3%, 90.0%, and 89.3% relative cell numbers, respectively, but it suddenly decreased to ∼73% for the 24 h/d treatment. Our results showed that the effects of tumor treating fields on tumor cells were higher than on normal cells for treatment duration of 3 to 12 h/d, but the difference became minimal for treatment duration of 24 h/d. The fractionated scheme, using tumor treating fields, reduced the treatment time while maintaining efficacy, suggesting that this method may be clinically applicable for cancer treatment.
Collapse
Affiliation(s)
- Yunhui Jo
- 1 Department of Bio-convergence Engineering, Korea University, Seoul, Korea.,2 Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Jiwon Sung
- 1 Department of Bio-convergence Engineering, Korea University, Seoul, Korea
| | - Hyesun Jeong
- 3 Department of Biomedical Science, Korea University, Seoul, Korea
| | - Sunghoi Hong
- 3 Department of Biomedical Science, Korea University, Seoul, Korea
| | - Youn Kyoung Jeong
- 4 Radiation Non-clinical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Eun Ho Kim
- 2 Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Myonggeun Yoon
- 1 Department of Bio-convergence Engineering, Korea University, Seoul, Korea
| |
Collapse
|
154
|
A theoretical study on real time monitoring of single cell mitosis with micro electrical impedance tomography. Biomed Microdevices 2019; 21:102. [PMID: 31768642 DOI: 10.1007/s10544-019-0452-3] [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] [Indexed: 10/25/2022]
Abstract
Real time monitoring of cell division, mitosis, at the single cell level, has value for many biomedical applications; such as developing optimal cancer treatments that target the cell division process. The goal of this theoretical study is to explore the feasibility of using Micro Electrical Impedance Tomography (MEIT) for real time monitoring of mitosis in a single cell, through imaging. MEIT employs a micro (single cell) scale electrode cage with electrodes placed around the cell. The electrodes deliver subsensory current and the consequential voltages on the electrodes are measured. An inverse image reconstruction algorithm uses the electric data from the electrodes to generate a map of electrical conductivity distribution in the chamber, which is the image. EIT is a well-known medical imaging technology that is simple to use but lacks good resolution. Therefore, it is not a-priori obvious that EIT has sufficient resolution to monitor single cell mitosis. To accomplish the goal of this study we have developed a mathematical model of MEIT of single cell mitosis, in which an in silico experiment provided the data for the MEIT image reconstruction. This theoretical study shows that MEIT can detect the outlines of the dividing cell during the various stages of mitosis (metaphase, anaphase and telophase) and, therefore, has potential as a technology for real time monitoring of single cell mitosis.
Collapse
|
155
|
Gentilal N, Salvador R, Miranda PC. Temperature control in TTFields therapy of GBM: impact on the duty cycle and tissue temperature. Phys Med Biol 2019; 64:225008. [PMID: 31671414 DOI: 10.1088/1361-6560/ab5323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In TTFields therapy, Optune® is used to deliver the electric field to the tumor via 4 transducer arrays. This device monitors the temperature of the transducers and reduces the current whenever a transducer reaches 41 °C. Our aim is to quantify Optune's duty cycle and to predict the steady-state temperature distribution in the head during GBM treatment. We used a realistic head model and the finite element method to solve Pennes equation and to simulate how Optune operates considering that current reduces to zero when the thermal limit is reached. The thermal impact was evaluated considering the maximum temperature reached by each tissue and using the CEM 43 °C metric. We observed that Optune switches the current on and off intermittently. In our model, one transducer reached the temperature limit quicker than the others and consequently it was the one that controlled current injection. This led to different duty cycles for the anterior-posterior and left-right array pairs. The thermal analysis indicated that the highest temperature in the model, 41.7 °C, was reached on the scalp under a transducer. However, TTFields may lead to significant changes only at the brain level such as BBB permeability increase, cerebral blood flow variation and changes in the concentration of some neurotransmitters. The duty cycle may be increased, e.g. by controlling the current at the transducer level. These predictions should be validated by comparison with experimental data and reconciled with the lack of evidence of thermal impact in clinical trials.
Collapse
Affiliation(s)
- Nichal Gentilal
- Institute of Biophysics and Biomedical Engineering, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal. Author to whom correspondence should be addressed
| | | | | |
Collapse
|
156
|
Investigation of uniform sized multicellular spheroids raised by microwell arrays after the combined treatment of electric field and anti-cancer drug. Biomed Microdevices 2019; 21:94. [DOI: 10.1007/s10544-019-0442-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
157
|
Pratiwi R, Antara NY, Fadliansyah LG, Ardiansyah SA, Nurhidayat L, Sholikhah EN, Sunarti S, Widyarini S, Fadhlurrahman AG, Fatmasari H, Tunjung WAS, Haryana SM, Alamsyah F, Taruno WP. CCL2 and IL18 expressions may associate with the anti-proliferative effect of noncontact electro capacitive cancer therapy in vivo. F1000Res 2019; 8:1770. [PMID: 32695310 PMCID: PMC7348523 DOI: 10.12688/f1000research.20727.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/14/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Noncontact Electro Capacitive Cancer Therapy (ECCT) is a novel treatment modality in cancer. Chemokine (C-C motif) ligand 2 (CCL2) has a major role in the outgrowth of metastatic breast cancer. Interleukin 18 (IL18) plays a role in macrophage alteration, which leads to excessive angiogenesis. This study aims to elaborate on the association of CCL2, IL18, IL23α, and TNF-α (tumor necrosis factor-alpha) expression with the anti-proliferative effect of ECCT in rat breast tumor tissue. Methods: Low intensity (18 Vpp) and intermediate frequency (150 kHz) alternating current-electric field (AC-EF) between two capacitive electrodes were exposed as external EF to a rat cage. Twenty-four rats were divided into four groups of six replicates. Breast tumor tissues were collected from 7, 12-dimethylbenz[a]anthracene (DMBA)-induced rats. Two groups were non DMBA-induced rats without ECCT exposure (NINT) and with (NIT). The other two groups were DMBA-induced rats without ECCT exposure (INT) and with (IT). Mammary glands and breast tumor tissues were collected from each group and preserved. Hematoxylin-eosin and immunohistochemistry staining were performed on paraffin sections of tissues using anti-PCNA, anti-ErbB2, anti-Caspase3, and anti-CD68. CCL2, IL18, IL23α, and TNF-α mRNA relative expressions were analyzed using qRT-PCR. Results: ECCT exposure may cause the reduction of PCNA protein expression as well as ErbB2 on breast tumor tissues, but it causes the increase of Caspase3 and macrophage CD68 protein. In rat breast tumor tissues of IT groups, the mRNA expression of CCL2 and IL18 are significantly down-regulated, in contrast with the up-regulated expression of these cytokines in tumor tissues of the INT group. IL23α and TNF- α expression remained similar in both groups. Conclusion: CCL2 and IL18 expressions have an association with the inhibition of breast tumor cell proliferation affected by ECCT exposure
Collapse
Affiliation(s)
- Rarastoeti Pratiwi
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia.,Graduate School of Biotechnology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Nyoman Yudi Antara
- Graduate School of Biotechnology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | | | | | - Luthfi Nurhidayat
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Eti Nurwening Sholikhah
- Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Sunarti Sunarti
- Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Sitarina Widyarini
- Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | | | - Hindana Fatmasari
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | | | - Sofia Mubarika Haryana
- Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Firman Alamsyah
- Center for Medical Physics and Cancer Research, Ctech Labs Edwar Technology, Tangerang, 15320, Indonesia
| | - Warsito Purwo Taruno
- Center for Medical Physics and Cancer Research, Ctech Labs Edwar Technology, Tangerang, 15320, Indonesia
| |
Collapse
|
158
|
Onken J, Goerling U, Heinrich M, Pleissner S, Krex D, Vajkoczy P, Misch M. Patient Reported Outcome (PRO) Among High-Grade Glioma Patients Receiving TTFields Treatment: A Two Center Observational Study. Front Neurol 2019; 10:1026. [PMID: 31681134 PMCID: PMC6797850 DOI: 10.3389/fneur.2019.01026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023] Open
Abstract
Study design: A two center, observational study. Introduction: Patient reported outcome (PRO) plays an increasingly important role in the evaluation of novel therapies for tumor patients. It has been shown that tumor treating fields (TTFields) in combination with standard therapy prolong survival in high-grade glioma (hgG) patients. But critics claim that TTFields significantly impacts patients' everyday life due to side effects and average daily time on therapy (18 h) in a patient population with very limited life expectancy and high symptom burden. However, very limited data exist on PRO for TTFields treatment. Methods: This two center, observational study describes PRO of 30 hgG patients receiving TTFields in combination with chemotherapy. We introduced a device-specific questionnaire (DSQ) addressing device-specific restrictions and impact on daily live after 2 months of therapy. Additionally following questionnaires were used: EORTC (European Organization for Research and Treatment of Cancer), QLQ-30 (Quality of life of cancer patients), QLQ BN20 (Quality of life brain cancer module), QLQ FA13 (Cancer-related fatigue), and SSUK-8 (social support). Results: Surveys have been completed by 91% of enrolled patients. EORTC QLQ-30 revealed better physical, emotional, and cognitive function than social and role function of study cohort. TTFields users reported frequently on positive social support and a low level of detrimental interactions. Seventy one percent of patients felt affected in daily life due to TTFields at least 2-3 times per week up to several times per day while maintaining high therapy compliance. Most frequent device-specific restrictions were duration of therapy (74%), size (66%), and weight (70%) of the device and changing time and bonding of the transducer arrays (66%, mean duration: 43.6 min). Restrictions on exercise of hobbies/work (63%/61%), body care (71%), and sexuality/relationship (64%) were most relevant. Seventy percent would recommend TTFields to others and 67% would reuse TTFields treatment again based on their current experience. Conclusion: The study shows that although TTFields treatment frequently affects everyday life in all aspects, therapy compliance was high and 67% of patients would reconsider TTFields for themselves. We propose that findings of PRO be taken into account for medical consultation about TTFields and in future device development to deliver high-value patient-centered care.
Collapse
Affiliation(s)
- Julia Onken
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin School of Integrative Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ute Goerling
- Psychooncology, Charité Comprehensive Cancer Center, Berlin, Germany
| | - Marcel Heinrich
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stephanie Pleissner
- Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Dietmar Krex
- Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Misch
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
159
|
Hershkovich HS, Urman N, Yesharim O, Naveh A, Bomzon Z. The dielectric properties of skin and their influence on the delivery of tumor treating fields to the torso: a study combining in vivo measurements with numerical simulations. ACTA ACUST UNITED AC 2019; 64:185014. [DOI: 10.1088/1361-6560/ab33c6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
160
|
Taphoorn MJB, Dirven L, Kanner AA, Lavy-Shahaf G, Weinberg U, Taillibert S, Toms SA, Honnorat J, Chen TC, Sroubek J, David C, Idbaih A, Easaw JC, Kim CY, Bruna J, Hottinger AF, Kew Y, Roth P, Desai R, Villano JL, Kirson ED, Ram Z, Stupp R. Influence of Treatment With Tumor-Treating Fields on Health-Related Quality of Life of Patients With Newly Diagnosed Glioblastoma: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol 2019; 4:495-504. [PMID: 29392280 DOI: 10.1001/jamaoncol.2017.5082] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Importance Tumor-treating fields (TTFields) therapy improves both progression-free and overall survival in patients with glioblastoma. There is a need to assess the influence of TTFields on patients' health-related quality of life (HRQoL). Objective To examine the association of TTFields therapy with progression-free survival and HRQoL among patients with glioblastoma. Design, Setting, and Participants This secondary analysis of EF-14, a phase 3 randomized clinical trial, compares TTFields and temozolomide or temozolomide alone in 695 patients with glioblastoma after completion of radiochemotherapy. Patients with glioblastoma were randomized 2:1 to combined treatment with TTFields and temozolomide or temozolomide alone. The study was conducted from July 2009 until November 2014, and patients were followed up through December 2016. Interventions Temozolomide, 150 to 200 mg/m2/d, was given for 5 days during each 28-day cycle. TTFields were delivered continuously via 4 transducer arrays placed on the shaved scalp of patients and were connected to a portable medical device. Main Outcomes and Measures Primary study end point was progression-free survival; HRQoL was a predefined secondary end point, measured with questionnaires at baseline and every 3 months thereafter. Mean changes from baseline scores were evaluated, as well as scores over time. Deterioration-free survival and time to deterioration were assessed for each of 9 preselected scales and items. Results Of the 695 patients in the study, 639 (91.9%) completed the baseline HRQoL questionnaire. Of these patients, 437 (68.4%) were men; mean (SD) age, 54.8 (11.5) years. Health-related quality of life did not differ significantly between treatment arms except for itchy skin. Deterioration-free survival was significantly longer with TTFields for global health (4.8 vs 3.3 months; P < .01); physical (5.1 vs 3.7 months; P < .01) and emotional functioning (5.3 vs 3.9 months; P < .01); pain (5.6 vs 3.6 months; P < .01); and leg weakness (5.6 vs 3.9 months; P < .01), likely related to improved progression-free survival. Time to deterioration, reflecting the influence of treatment, did not differ significantly except for itchy skin (TTFields worse; 8.2 vs 14.4 months; P < .001) and pain (TTFields improved; 13.4 vs 12.1 months; P < .01). Role, social, and physical functioning were not affected by TTFields. Conclusions and Relevance The addition of TTFields to standard treatment with temozolomide for patients with glioblastoma results in improved survival without a negative influence on HRQoL except for more itchy skin, an expected consequence from the transducer arrays. Trial Registration clinicaltrials.gov Identifier: NCT00916409.
Collapse
Affiliation(s)
- Martin J B Taphoorn
- Department of Neurology, Haaglanden Medical Center, The Hague, The Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Linda Dirven
- Department of Neurology, Haaglanden Medical Center, The Hague, The Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew A Kanner
- Department of Neurosurgery, Rabin Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Uri Weinberg
- Research and Development, Novocure, Haifa, Switzerland
| | - Sophie Taillibert
- Department of Neurology 2, Salpêtrière University Hospital, Assistance Public Hôpitaux de Paris, L'Université Pierre et Marie Curie University, Paris VI University, Paris, France
| | - Steven A Toms
- Department of Neurosurgery, Geisinger Medical Center, Danville, Pennsylvania
| | - Jerome Honnorat
- Department of Neuro-oncology, Hospices Civils de Lyon, University Claude Bernard Lyon, Lyon, France
| | - Thomas C Chen
- Department of Neurosurgery, University of Southern California, Los Angeles
| | - Jan Sroubek
- Department of Neurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - Carlos David
- Department of Neurosurgery, Lahey Clinic, Burlington, Massachusetts
| | - Ahmed Idbaih
- Department of Neurology 2, Salpêtrière University Hospital, Assistance Public Hôpitaux de Paris, L'Université Pierre et Marie Curie University, Paris VI University, Paris, France
| | - Jacob C Easaw
- Department of Medical Oncology, Cross Cancer Institute, Edmonton, California
| | - Chae-Yong Kim
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Bundang, Korea
| | - Jordi Bruna
- Department of Neurology, Hospital Universitari Bellvitge, Barcelona, Spain
| | - Andreas F Hottinger
- Department of Medical Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Yvonne Kew
- Clinical Neuro-Oncology Research Program, Department of Internal Medicine, Methodist Hospital, Houston, Texas
| | - Patrick Roth
- Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Rajiv Desai
- Neurosurgery and Spine Association, Maine Medical Center, Scarborough, Maine
| | - John L Villano
- Clinical Neuro-Oncology Research Program, Department of Internal Medicine, University of Kentucky Medical Center, Lexington
| | | | - Zvi Ram
- Department of Neurosurgery, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Roger Stupp
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Northwestern Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
161
|
Mattsson MO, Simkó M. Emerging medical applications based on non-ionizing electromagnetic fields from 0 Hz to 10 THz. MEDICAL DEVICES-EVIDENCE AND RESEARCH 2019; 12:347-368. [PMID: 31565000 PMCID: PMC6746309 DOI: 10.2147/mder.s214152] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/23/2019] [Indexed: 12/13/2022] Open
Abstract
The potential for using non-ionizing electromagnetic fields (EMF; at frequencies from 0 Hz up to the THz range) for medical purposes has been of interest since many decades. A number of established and familiar methods are in use all over the world. This review, however, provides an overview of applications that already play some clinical role or are in earlier stages of development. The covered methods include modalities used for bone healing, cancer treatment, neurological conditions, and diathermy. In addition, certain other potential clinical areas are touched upon. Most of the reviewed technologies deal with therapy, whereas just a few diagnostic approaches are mentioned. None of the discussed methods are having such a strong impact in their field of use that they would be expected to replace conventional methods. Partly this is due to a knowledge base that lacks mechanistic explanations for EMF effects at low-intensity levels, which often are used in the applications. Thus, the possible optimal use of EMF approaches is restricted. Other reasons for the limited impact include a scarcity of well-performed randomized clinical trials that convincingly show the efficacy of the methods and that standardized user protocols are mostly lacking. Presently, it seems that some EMF-based methods can have a niche role in treatment and diagnostics of certain conditions, mostly as a complement to or in combination with other, more established, methods. Further development and a stronger impact of these technologies need a better understanding of the interaction mechanisms between EMF and biological systems at lower intensity levels. The importance of the different physical parameters of the EMF exposure needs also further investigations.
Collapse
Affiliation(s)
- Mats-Olof Mattsson
- SciProof International AB, Östersund, Sweden
- Strömstad Akademi, Institute for Advanced Studies, Strömstad, Sweden
| | - Myrtill Simkó
- SciProof International AB, Östersund, Sweden
- Strömstad Akademi, Institute for Advanced Studies, Strömstad, Sweden
| |
Collapse
|
162
|
Hernández-Bule ML, Medel E, Colastra C, Roldán R, Úbeda A. Response of neuroblastoma cells to RF currents as a function of the signal frequency. BMC Cancer 2019; 19:889. [PMID: 31488097 PMCID: PMC6728948 DOI: 10.1186/s12885-019-6090-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Capacitive-resistive electric transfer (CRET) is a non-invasive therapeutic strategy that applies radiofrequency electric currents within the 400-600 kHz range to tissue repair and regeneration. Previous studies by our group have shown that 48 h of intermittent exposure to a 570 kHz CRET signal at a subthermal density of 50 μA/mm2 causes significant changes in the expression and activation of cell cycle control proteins, leading to cycle arrest in human cancer cell cultures. The present study investigates the relevance of the signal frequency in the response of the human neuroblastoma cell line NB69 to subthermal electric treatment with four different signal frequency currents within the 350-650 kHz range. METHODS Trypan blue assay, flow cytometry, immunofluorescence and immunoblot were used to study the effects of subthermal CRET currents on cell viability, cell cycle progression and the expression of several marker proteins involved in NB69 cell death and proliferation. RESULTS The results reveal that among the frequencies tested, only a 448 kHz signal elicited both proapoptotic and antiproliferative, statistically significant responses. The apoptotic effect would be due, at least in part, to significant changes induced by the 448 kHz signal in the expression of p53, Bax and caspase-3. The cytostatic response was preceded by alterations in the kinetics of the cell cycle and in the expression of proteins p-ERK1/2, cyclin D1 and p27, which is consistent with a potential involvement of the EGF receptor in electrically induced changes in the ERK1/2 pathway. This receives additional support from results indicating that the proapototic and antiproliferative responses to CRET can be transiently blocked when the electric stimulus is applied in the presence of PD98059, a chemical inhibitor of the ERK1/2 pathway. CONCLUSION The understanding of the mechanisms underlying the ability of slowing down cancer cell growth through electrically-induced changes in the expression of proteins involved in the control of cell proliferation and apoptosis might afford new insights in the field of oncology.
Collapse
Affiliation(s)
- María Luisa Hernández-Bule
- BEM-Research Service, Ramón y Cajal University Hospital - IRYCIS, Ctra. Colmenar Viejo km 9-100, 28034, Madrid, Spain.
| | - Enrique Medel
- BEM-Research Service, Ramón y Cajal University Hospital - IRYCIS, Ctra. Colmenar Viejo km 9-100, 28034, Madrid, Spain
| | - Clara Colastra
- BEM-Research Service, Ramón y Cajal University Hospital - IRYCIS, Ctra. Colmenar Viejo km 9-100, 28034, Madrid, Spain
| | - Raquel Roldán
- BEM-Research Service, Ramón y Cajal University Hospital - IRYCIS, Ctra. Colmenar Viejo km 9-100, 28034, Madrid, Spain
| | - Alejandro Úbeda
- BEM-Research Service, Ramón y Cajal University Hospital - IRYCIS, Ctra. Colmenar Viejo km 9-100, 28034, Madrid, Spain
| |
Collapse
|
163
|
Stachelek GC, Grimm J, Moore J, Huang E, Spoleti N, Redmond KJ, Lim M, Bettegowda C, Kleinberg L. Tumor-Treating Field Arrays Do Not Reduce Target Volume Coverage for Glioblastoma Radiation Therapy. Adv Radiat Oncol 2019; 5:62-69. [PMID: 32051891 PMCID: PMC7004938 DOI: 10.1016/j.adro.2019.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/11/2019] [Accepted: 08/14/2019] [Indexed: 11/30/2022] Open
Abstract
Purpose To inform development of procedures for using tumor-treating field arrays (TTFields) during glioblastoma radiation therapy by determining whether the placement and repositioning of arrays affects target volume coverage and cranial skin dose. Methods and Materials Radiation plans from 10 consecutive patients treated for glioblastoma were copied to a cranial phantom and reoptimized for phantom anatomy. Dose distributions were then recalculated on 3 additional computed tomographic scans of the phantom with the TTFields electrode arrays placed over distinct locations on the phantom scalp to compare planning target volume (PTV) coverage and skin dose with and without TTFields in place in varying positions. Percent depth dose curves were also measured for radiation beams passing through the electrodes and compared with commonly used bolus material. Results The presence of TTFields arrays decreased PTV V97% and D97% by as much as 1.7% and 2.7%, respectively, for a single array position, but this decrease was mitigated by array repositioning. On averaging the 3 array positions, there was no statistically significant difference in any dosimetric parameter of PTV coverage (V95-97%, D95-97%) across all cases compared with no array. Mean increases in skin D1cc and D20cc of 3.1% were calculated for the cohort. Surface dose for TTFields electrodes was less than that with a 5-mm superflab bolus. Conclusions Our work demonstrates that placement of TTFields arrays does not significantly affect target volume coverage. We show that repositioning of TTFields arrays, as is required in clinical use, further minimizes any dosimetric changes and eliminates the need for replanning when arrays are moved. A slight, expected bolus effect is observed, but the calculated increases in skin dose are not clinically significant. These data support the development of clinical trials to assess the safety and efficacy of combining concurrent chemoradiotherapy with TTFields therapy for glioblastoma.
Collapse
Affiliation(s)
- Gregory C Stachelek
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, Maryland
| | - Jimm Grimm
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, Maryland
| | - Joseph Moore
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, Maryland
| | - Ellen Huang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, Maryland
| | - Nicholas Spoleti
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, Maryland
| | - Kristin J Redmond
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, Maryland
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, Maryland
| | - Lawrence Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
164
|
Wang Y, Pandey M, Ballo MT. Integration of Tumor-Treating Fields into the Multidisciplinary Management of Patients with Solid Malignancies. Oncologist 2019; 24:e1426-e1436. [PMID: 31444292 PMCID: PMC6975944 DOI: 10.1634/theoncologist.2017-0603] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/10/2019] [Indexed: 12/24/2022] Open
Abstract
Tumor treating fields, a noninvasive cancer treatment using low intensity alternating electric fields, offers clinical opportunities with unique challenges. This review focuses on the mechanism of action of this treatment, the known pre‐clinical and clinical experience, and the practical issues surrounding its use in the multidisciplinary management of patients with solid malignancies. Tumor‐treating fields (TTFields) are a noninvasive antimitotic cancer treatment consisting of low‐intensity alternating electric fields delivered to the tumor or tumor bed via externally applied transducer arrays. In multiple in vitro and in vivo cancer cell lines, TTFields therapy inhibits cell proliferation, disrupts cell division, interferes with cell migration and invasion, and reduces DNA repair. Human trials in patients with primary glioblastoma showed an improvement in overall survival, and trials in patients with unresectable malignant pleural mesothelioma showed favorable outcomes compared with historical control. This led to U.S. Food and Drug Administration approval in both clinical situations, paving the way for development of trials investigating TTFields in other malignancies. Although these trials are ongoing, the existing evidence suggests that TTFields have activity outside of neuro‐oncology, and further study into the mechanism of action and clinical activity is required. In addition, because TTFields are a previously unrecognized antimitotic therapy with a unique mode of delivery, the oncological community must address obstacles to widespread patient and provider acceptance. TTFields will likely join surgery, systemic therapy, and radiation therapy as a component of multimodality management of patients with solid malignancies. Implications for Practice. Tumor‐treating fields (TTFields) exhibit a broad range of antitumor activities. Clinically, they improve overall survival for patients with newly diagnosed glioblastoma. The emergence of TTFields has changed the treatment regimen for glioblastoma. Clinicians need to understand the practical issues surrounding its use in the multidisciplinary management of patients with glioblastoma. With ongoing clinical trials, TTFields likely will become another treatment modality for solid malignancies.
Collapse
Affiliation(s)
- Yuefeng Wang
- Department of Radiation Oncology, West Cancer Center and Research Institute, Memphis, Tennessee, USA
| | - Manjari Pandey
- Department of Hematology/Oncology, West Cancer Center and Research Institute, Memphis, Tennessee, USA
| | - Matthew T Ballo
- Department of Radiation Oncology, West Cancer Center and Research Institute, Memphis, Tennessee, USA
| |
Collapse
|
165
|
Najberg M, Haji Mansor M, Boury F, Alvarez-Lorenzo C, Garcion E. Reversing the Tumor Target: Establishment of a Tumor Trap. Front Pharmacol 2019; 10:887. [PMID: 31456685 PMCID: PMC6699082 DOI: 10.3389/fphar.2019.00887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Despite the tremendous progress made in the field of cancer therapy in recent years, certain solid tumors still cannot be successfully treated. Alongside classical treatments in the form of chemotherapy and/or radiotherapy, targeted treatments such as immunotherapy that cause fewer side effects emerge as new options in the clinics. However, these alternative treatments may not be useful for treating all types of cancers, especially for killing infiltrative and circulating tumor cells (CTCs). Recent advances pursue the trapping of these cancer cells within a confined area to facilitate their removal for therapeutic and diagnostic purposes. A good understanding of the mechanisms behind tumor cell migration may drive the design of traps that mimic natural tumor niches and guide the movement of the cancer cells. To bring this trapping idea into reality, strong efforts are being made to create structured materials that imitate myelinated fibers, blood vessels, or pre-metastatic niches and incorporate chemical cues such as chemoattractants or adhesive proteins. In this review, the different strategies used (or could be used) to trap tumor cells are described, and relevant examples of their performance are analyzed.
Collapse
Affiliation(s)
- Mathie Najberg
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R + D Pharma Group (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Muhammad Haji Mansor
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- Center for Education and Research on Macromolecules (CERM), Université de Liège, Liège, Belgium
| | - Frank Boury
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R + D Pharma Group (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Emmanuel Garcion
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| |
Collapse
|
166
|
Ballo MT, Urman N, Lavy-Shahaf G, Grewal J, Bomzon Z, Toms S. Correlation of Tumor Treating Fields Dosimetry to Survival Outcomes in Newly Diagnosed Glioblastoma: A Large-Scale Numerical Simulation-Based Analysis of Data from the Phase 3 EF-14 Randomized Trial. Int J Radiat Oncol Biol Phys 2019; 104:1106-1113. [DOI: 10.1016/j.ijrobp.2019.04.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/16/2019] [Accepted: 04/14/2019] [Indexed: 10/27/2022]
|
167
|
Tumour-treating fields (TTFields): Investigations on the mechanism of action by electromagnetic exposure of cells in telophase/cytokinesis. Sci Rep 2019; 9:7362. [PMID: 31089145 PMCID: PMC6517379 DOI: 10.1038/s41598-019-43621-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/26/2019] [Indexed: 11/25/2022] Open
Abstract
Tumour-treating fields (TTFields) use alternating electric fields which interfere with dividing cells, thereby reducing tumour growth. Previous reports suggest that electrical forces on cell structure proteins interfered with the chromosome separation during mitosis and induced apoptosis. In the present report we evaluate electromagnetic exposure of cells in telophase/cytokinesis in order to further analyse the mechanism of action on cells. We performed numerical electromagnetic simulations to analyse the field distribution in a cell during different mitotic phases. Based thereon, we developed an electric lumped element model of the mitotic cell. Both the electromagnetic simulation and the lumped element model predict a local increase of the specific absorption rate (SAR) as a measure of the electromagnetically induced power absorption density at the mitotic furrow which may help to explain the anti-proliferative effect. In accordance with other reports, cell culture experiments confirmed that TTFields reduce the proliferation of different glioma cell lines in a field strength- and frequency-dependent manner. Furthermore, we found an additional dependence on the commutation time of the electrical fields. The report gives new insights into TTFields’ anti-proliferative effect on tumours, which could help to improve future TTFields application systems.
Collapse
|
168
|
Huang CH, Lei KF, Tsang NM. Apoptosis and cell cycle arrest of hepatocellular carcinoma spheroids treated by an alternating electric field. Biotechnol Prog 2019; 35:e2787. [DOI: 10.1002/btpr.2787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/17/2018] [Accepted: 02/05/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Chun-Hao Huang
- Program in Biomedical Engineering; College of Engineering, Chang Gung University; Taoyuan Taiwan
| | - Kin Fong Lei
- Graduate Institute of Biomedical Engineering, Chang Gung University; Taoyuan Taiwan
- Department of Radiation Oncology; Chang Gung Memorial Hospital; Linkou Taiwan
| | - Ngan-Ming Tsang
- Department of Radiation Oncology; Chang Gung Memorial Hospital; Linkou Taiwan
- Department of Traditional Chinese Medicine; Chang Gung University; Taoyuan Taiwan
| |
Collapse
|
169
|
|
170
|
|
171
|
Alternating Electric Fields (TTFields) Activate Ca v1.2 Channels in Human Glioblastoma Cells. Cancers (Basel) 2019; 11:cancers11010110. [PMID: 30669316 PMCID: PMC6356873 DOI: 10.3390/cancers11010110] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/16/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor treating fields (TTFields) represent a novel FDA-approved treatment modality for patients with newly diagnosed or recurrent glioblastoma multiforme. This therapy applies intermediate frequency alternating electric fields with low intensity to the tumor volume by the use of non-invasive transducer electrode arrays. Mechanistically, TTFields have been proposed to impair formation of the mitotic spindle apparatus and cytokinesis. In order to identify further potential molecular targets, here the effects of TTFields on Ca2+-signaling, ion channel activity in the plasma membrane, cell cycle, cell death, and clonogenic survival were tested in two human glioblastoma cell lines in vitro by fura-2 Ca2+ imaging, patch-clamp cell-attached recordings, flow cytometry and pre-plated colony formation assay. In addition, the expression of voltage-gated Ca2+ (Cav) channels was determined by real-time RT-PCR and their significance for the cellular TTFields response defined by knock-down and pharmacological blockade. As a result, TTFields stimulated in a cell line-dependent manner a Cav1.2-mediated Ca2+ entry, G1 or S phase cell cycle arrest, breakdown of the inner mitochondrial membrane potential and DNA degradation, and/or decline of clonogenic survival suggesting a tumoricidal action of TTFields. Moreover, inhibition of Cav1.2 by benidipine aggravated in one glioblastoma line the TTFields effects suggesting that Cav1.2-triggered signaling contributes to cellular TTFields stress response. In conclusion, the present study identified Cav1.2 channels as TTFields target in the plasma membrane and provides the rationale to combine TTFields therapy with Ca2+ antagonists that are already in clinical use.
Collapse
|
172
|
Toms SA, Kim CY, Nicholas G, Ram Z. Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF-14 phase III trial. J Neurooncol 2019; 141:467-473. [PMID: 30506499 PMCID: PMC6342854 DOI: 10.1007/s11060-018-03057-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/21/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Tumor treating fields (TTFields) is a non-invasive, antimitotic therapy. In the EF-14 phase 3 trial in newly diagnosed glioblastoma, TTFields plus temozolomide (TTFields/TMZ) improved progression free (PFS) and overall survival (OS) versus TMZ alone. Previous data indicate a ≥ 75% daily compliance improves outcomes. We analyzed compliance data from TTFields/TMZ patients in the EF-14 study to correlate TTFields compliance with PFS and OS and identify potential lower boundary for compliance with improved clinical outcomes. METHODS Compliance was assessed by usage data from the NovoTTF-100A device and calculated as percentage per month of TTFields delivery. TTFields/TMZ patients were segregated into subgroups by percent monthly compliance. A Cox proportional hazard model controlled for sex, extent of resection, MGMT methylation status, age, region, and performance status was used to investigate the effect of compliance on PFS and OS. RESULTS A threshold value of 50% compliance with TTFields/TMZ improved PFS (HR 0.70, 95% CI 0.47-1.05) and OS (HR 0.67, 95% CI 0.45-0.99) versus TMZ alone with improved outcome as compliance increased. At compliance > 90%, median survival was 24.9 months (28.7 months from diagnosis) and 5-year survival rate was 29.3%. Compliance was independent of gender, extent of resection, MGMT methylation status, age, region and performance status (HR 0.78; p = 0.031; OS at compliance ≥ 75% vs. < 75%). CONCLUSION A compliance threshold of 50% with TTFields/TMZ correlated with significantly improved OS and PFS versus TMZ alone. Patients with compliance > 90% showed extended median and 5-year survival rates. Increased compliance with TTFields therapy is independently prognostic for improved survival in glioblastoma.
Collapse
Affiliation(s)
- S A Toms
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - C Y Kim
- Seoul National University, Bundang, South Korea
| | - G Nicholas
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Z Ram
- Tel Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
173
|
Rivera F, Benavides M, Gallego J, Guillen-Ponce C, Lopez-Martin J, Küng M. Tumor treating fields in combination with gemcitabine or gemcitabine plus nab-paclitaxel in pancreatic cancer: Results of the PANOVA phase 2 study. Pancreatology 2019; 19:64-72. [PMID: 30396819 DOI: 10.1016/j.pan.2018.10.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 09/18/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Tumor Treating Fields (TTFields), low intensity alternating electric fields with antimitotic activity, have demonstrated survival benefit in patients with glioblastoma. This phase 2 PANOVA study was conducted to examine the combination of TTFields plus chemotherapy in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS Forty patients with newly-diagnosed, locally advanced or metastatic PDAC received continuous TTFields (150 KHz for ≥18 h/day) plus gemcitabine (1000 mg/m2), or gemcitabine plus nab-paclitaxel (125 mg/m2). The primary endpoint was safety and secondary endpoints included compliance to TTFields, progression-free survival (PFS), and overall survival (OS). RESULTS Seventeen patients (85%) in each cohort reported Grade ≥3 adverse events (AEs). No increase in serious AEs (SAEs) was observed compared to that anticipated with systemic chemotherapy alone. Twenty-one patients reported TTFields-related skin toxicity, of which 7 were Grade 3; all resolved following temporary reduction of daily TTFields usage. No TTFields-related SAEs were reported. Compliance to TTFields was 68-78% of the recommended average daily use in both cohorts. Median PFS was 8.3 months (95% CI 4.3, 10.3) and median OS was 14.9 months (95% CI 6.2, NA) in the TTFields + gemcitabine cohort. In the TTFields + gemcitabine + nab-paclitaxel cohort, the median PFS was 12.7 months (95% CI 5.4, NA); median OS has not been reached. CONCLUSION The PANOVA trial demonstrated that the combination of TTFields and systemic chemotherapy is safe and tolerable in patients with advanced PDAC. Based on the safety and preliminary efficacy results of this phase 2 study, a randomized phase 3 study (PANOVA-3) is underway.
Collapse
Affiliation(s)
- Fernando Rivera
- Hospital Universitario Marqués de Valdecilla, Santander, Spain.
| | - Manuel Benavides
- Hospital Universitario Regional y Virgen de la Victoria, Andalucia, Spain.
| | - Javier Gallego
- Plazas Hospital General Universitario de Elche, Elche, Spain.
| | | | | | - Marc Küng
- Hôpital Fribourgeois, Fribourg, Switzerland.
| |
Collapse
|
174
|
Chang E, Patel CB, Pohling C, Young C, Song J, Flores TA, Zeng Y, Joubert LM, Arami H, Natarajan A, Sinclair R, Gambhir SS. Tumor treating fields increases membrane permeability in glioblastoma cells. Cell Death Discov 2018; 4:113. [PMID: 30534421 PMCID: PMC6281619 DOI: 10.1038/s41420-018-0130-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma is the most common yet most lethal of primary brain cancers with a one-year post-diagnosis survival rate of 65% and a five-year survival rate of barely 5%. Recently the U.S. Food and Drug Administration approved a novel fourth approach (in addition to surgery, radiation therapy, and chemotherapy) to treating glioblastoma; namely, tumor treating fields (TTFields). TTFields involves the delivery of alternating electric fields to the tumor but its mechanisms of action are not fully understood. Current theories involve TTFields disrupting mitosis due to interference with proper mitotic spindle assembly. We show that TTFields also alters cellular membrane structure thus rendering it more permeant to chemotherapeutics. Increased membrane permeability through the imposition of TTFields was shown by several approaches. For example, increased permeability was indicated through increased bioluminescence with TTFields exposure or with the increased binding and ingress of membrane-associating reagents such as Dextran-FITC or ethidium D or with the demonstration by scanning electron microscopy of augmented number and sizes of holes on the cellular membrane. Further investigations showed that increases in bioluminescence and membrane hole production with TTFields exposure disappeared by 24 h after cessation of alternating electric fields thus demonstrating that this phenomenom is reversible. Preliminary investigations showed that TTFields did not induce membrane holes in normal human fibroblasts thus suggesting that the phenomenom was specific to cancer cells. With TTFields, we present evidence showing augmented membrane accessibility by compounds such as 5-aminolevulinic acid, a reagent used intraoperatively to delineate tumor from normal tissue in glioblastoma patients. In addition, this mechanism helps to explain previous reports of additive and synergistic effects between TTFields and other chemotherapies. These findings have implications for the design of combination therapies in glioblastoma and other cancers and may significantly alter standard of care strategies for these diseases.
Collapse
Affiliation(s)
- Edwin Chang
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Chirag B. Patel
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
- Division of Neuro-Oncology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Christoph Pohling
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Caroline Young
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Jonathan Song
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Thomas Anthony Flores
- Department of Applied Physics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Yitian Zeng
- Department of Materials Science & Engineering, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Lydia-Marie Joubert
- Electron Microscopy Unit, Stellenbosch University, Stellenbosch, South Africa
| | - Hamed Arami
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
| | - Robert Sinclair
- Department of Materials Science & Engineering, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Sanjiv S. Gambhir
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, Room E150, 318 Campus Drive West, Stanford, CA 94305 USA
- Department of Materials Science & Engineering, Stanford University School of Medicine, Stanford, CA 94305 USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305 USA
| |
Collapse
|
175
|
Branter J, Basu S, Smith S. Tumour treating fields in a combinational therapeutic approach. Oncotarget 2018; 9:36631-36644. [PMID: 30564303 PMCID: PMC6290966 DOI: 10.18632/oncotarget.26344] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 10/24/2018] [Indexed: 12/15/2022] Open
Abstract
The standard of care for patients with newly diagnosed Glioblastoma multiforme (GBM) has remained unchanged since 2005, with patients undergoing maximal surgical resection, followed by radiotherapy plus concomitant and maintenance Temozolomide. More recently, Tumour treating fields (TTFields) therapy has become FDA approved for adult recurrent and adult newly-diagnosed GBM following the EF-11 and EF-14 trials, respectively. TTFields is a non-invasive anticancer treatment which utilizes medium frequency alternating electric fields to target actively dividing cancerous cells. TTFields selectively targets cells within mitosis through interacting with key mitotic proteins to cause mitotic arrest and cell death. TTFields therapy presents itself as a candidate for the combinational therapy route due to the lack of overlapping toxicities associated with electric fields. Here we review current literature pertaining to TTFields in combination with alkylating agents, radiation, anti-angiogenics, mitotic inhibitors, immunotherapies, and also with novel agents. This review highlights the observed synergistic and additive effects of combining TTFields with various other therapies, as well highlighting the strategies relating to combinations with electric fields.
Collapse
Affiliation(s)
- Joshua Branter
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Surajit Basu
- Queen's Medical Centre, Department of Neurosurgery, Nottingham, UK
| | - Stuart Smith
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
176
|
Jo Y, Kim EH, Sai S, Kim JS, Cho JM, Kim H, Baek JH, Kim JY, Hwang SG, Yoon M. Functional Biological Activity of Sorafenib as a Tumor-Treating Field Sensitizer for Glioblastoma Therapy. Int J Mol Sci 2018; 19:E3684. [PMID: 30469352 PMCID: PMC6274791 DOI: 10.3390/ijms19113684] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/09/2018] [Accepted: 11/16/2018] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma, the most common primary brain tumor in adults, is an incurable malignancy with poor short-term survival and is typically treated with radiotherapy along with temozolomide. While the development of tumor-treating fields (TTFields), electric fields with alternating low and intermediate intensity has facilitated glioblastoma treatment, clinical outcomes of TTFields are reportedly inconsistent. However, combinatorial administration of chemotherapy with TTFields has proven effective for glioblastoma patients. Sorafenib, an anti-proliferative and apoptogenic agent, is used as first-line treatment for glioblastoma. This study aimed to investigate the effect of sorafenib on TTFields-induced anti-tumor and anti-angiogenesis responses in glioblastoma cells in vitro and in vivo. Sorafenib sensitized glioblastoma cells to TTFields, as evident from significantly decreased post-TTFields cell viability (p < 0.05), and combinatorial treatment with sorafenib and TTFields accelerated apoptosis via reactive oxygen species (ROS) generation, as evident from Poly (ADP-ribose) polymerase (PARP) cleavage. Furthermore, use of sorafenib plus TTFields increased autophagy, as evident from LC3 upregulation and autophagic vacuole formation. Cell cycle markers accumulated, and cells underwent a G2/M arrest, with an increased G0/G1 cell ratio. In addition, the combinatorial treatment significantly inhibited tumor cell motility and invasiveness, and angiogenesis. Our results suggest that combination therapy with sorafenib and TTFields is slightly better than each individual therapy and could potentially be used to treat glioblastoma in clinic, which requires further studies.
Collapse
Affiliation(s)
- Yunhui Jo
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
- Department of Bio-Convergence Engineering, Korea University, Seoul 02842, Korea.
| | - Eun Ho Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Sei Sai
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Chiba 263-0024, Japan.
| | - Jin Su Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Jae-Min Cho
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Hyeongi Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Jeong-Hwa Baek
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Jeong-Yub Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea.
| | - Myonggeun Yoon
- Department of Bio-Convergence Engineering, Korea University, Seoul 02842, Korea.
| |
Collapse
|
177
|
Proliferation arrest, selectivity, and chemosensitivity enhancement of cancer cells treated by a low-intensity alternating electric field. Biomed Microdevices 2018; 20:90. [DOI: 10.1007/s10544-018-0339-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
178
|
Shteingauz A, Porat Y, Voloshin T, Schneiderman RS, Munster M, Zeevi E, Kaynan N, Gotlib K, Giladi M, Kirson ED, Weinberg U, Kinzel A, Palti Y. AMPK-dependent autophagy upregulation serves as a survival mechanism in response to Tumor Treating Fields (TTFields). Cell Death Dis 2018; 9:1074. [PMID: 30341282 PMCID: PMC6195570 DOI: 10.1038/s41419-018-1085-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 01/04/2023]
Abstract
Tumor Treating Fields (TTFields), an approved treatment modality for glioblastoma, are delivered via non-invasive application of low-intensity, intermediate-frequency, alternating electric fields. TTFields application leads to abnormal mitosis, aneuploidy, and increased cell granularity, which are often associated with enhancement of autophagy. In this work, we evaluated whether TTFields effected the regulation of autophagy in glioma cells. We found that autophagy is upregulated in glioma cells treated with TTFields as demonstrated by immunoblot analysis of the lipidated microtubule-associated protein light chain 3 (LC3-II). Fluorescence and transmission electron microscopy demonstrated the presence of LC3 puncta and typical autophagosome-like structures in TTFields-treated cells. Utilizing time-lapse microscopy, we found that the significant increase in the formation of LC3 puncta was specific to cells that divided during TTFields application. Evaluation of selected cell stress parameters revealed an increase in the expression of the endoplasmic reticulum (ER) stress marker GRP78 and decreased intracellular ATP levels, both of which are indicative of increased proteotoxic stress. Pathway analysis demonstrated that TTFields-induced upregulation of autophagy is dependent on AMP-activated protein kinase (AMPK) activation. Depletion of AMPK or autophagy-related protein 7 (ATG7) inhibited the upregulation of autophagy in response to TTFields, as well as sensitized cells to the treatment, suggesting that cancer cells utilize autophagy as a resistance mechanism to TTFields. Combining TTFields with the autophagy inhibitor chloroquine (CQ) resulted in a significant dose-dependent reduction in cell growth compared with either TTFields or CQ alone. These results suggest that dividing cells upregulate autophagy in response to aneuploidy and ER stress induced by TTFields, and that AMPK serves as a key regulator of this process.
Collapse
|
179
|
Selective toxicity of tumor treating fields to melanoma: an in vitro and in vivo study. Cell Death Discov 2018; 4:46. [PMID: 30302280 PMCID: PMC6170382 DOI: 10.1038/s41420-018-0106-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/07/2018] [Accepted: 07/10/2018] [Indexed: 11/08/2022] Open
Abstract
Tumor treating fields (TTFs) are a newly developed cancer therapy technology using an alternating electric field that may be a possible candidate for overcoming the limitations of conventional treatment methods currently used in cancer treatment. Although clinical results using TTFs appear promising, concerns regarding side effects must be clarified to demonstrate the effectiveness of this treatment method. To investigate the side effects of TTF treatment, the damage to normal cell lines and normal tissue of a mouse model was compared with the damage to tumor cells and tumors in a mouse model after TTF treatment. No serious damage was found in the normal cells and normal tissues of the mouse model, suggesting that the side effects of TTF treatment may not be serious. Our evidence based on in vitro and in vivo experiments suggests that TTF may cause selective damage to cancer cells, further demonstrating the potential of TTF as an attractive alternative to conventional cancer treatment modalities.
Collapse
|
180
|
Diaz P, Horne E, Xu C, Hamel E, Wagenbach M, Petrov RR, Uhlenbruck B, Haas B, Hothi P, Wordeman L, Gussio R, Stella N. Modified carbazoles destabilize microtubules and kill glioblastoma multiform cells. Eur J Med Chem 2018; 159:74-89. [PMID: 30268825 DOI: 10.1016/j.ejmech.2018.09.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/07/2018] [Accepted: 09/09/2018] [Indexed: 11/26/2022]
Abstract
Small molecules that target microtubules (MTs) represent promising therapeutics to treat certain types of cancer, including glioblastoma multiform (GBM). We synthesized modified carbazoles and evaluated their antitumor activity in GBM cells in culture. Modified carbazoles with an ethyl moiety linked to the nitrogen of the carbazole and a carbonyl moiety linked to distinct biaromatic rings exhibited remarkably different killing activities in human GBM cell lines and patient-derived GBM cells, with IC50 values from 67 to >10,000 nM. Measures of the activity of modified carbazoles with tubulin and microtubules coupled to molecular docking studies show that these compounds bind to the colchicine site of tubulin in a unique low interaction space that inhibits tubulin assembly. The modified carbazoles reported here represent novel chemical tools to better understand how small molecules disrupt MT functions and kill devastating cancers such as GBM.
Collapse
Affiliation(s)
- Philippe Diaz
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA; DermaXon LLC, 32 Campus Drive, Missoula, MT, 59812, USA.
| | - Eric Horne
- Stella Therapeutics, Inc., Pacific Northwest Research Institute, 720 Broadway, Seattle, WA, 98122, USA
| | - Cong Xu
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Ernest Hamel
- Screening Technologies Branch (EH) and Computational Drug Development Group (RG), Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Michael Wagenbach
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Ravil R Petrov
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
| | - Benjamin Uhlenbruck
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
| | - Brian Haas
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Parvinder Hothi
- Ivy Center for Advance Brain Tumor Treatment, Swedish Neuroscience Institute, 550 17th Ave, Seattle, WA, 98122, USA
| | - Linda Wordeman
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Rick Gussio
- Screening Technologies Branch (EH) and Computational Drug Development Group (RG), Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Nephi Stella
- Stella Therapeutics, Inc., Pacific Northwest Research Institute, 720 Broadway, Seattle, WA, 98122, USA; Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
181
|
Vergote I, von Moos R, Manso L, Van Nieuwenhuysen E, Concin N, Sessa C. Tumor Treating Fields in combination with paclitaxel in recurrent ovarian carcinoma: Results of the INNOVATE pilot study. Gynecol Oncol 2018; 150:471-477. [DOI: 10.1016/j.ygyno.2018.07.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/18/2018] [Accepted: 07/22/2018] [Indexed: 12/31/2022]
|
182
|
Huang CH, Lei KF, Tsang NM. Dissociated effect and Chemosensitive enhancement of tumor spheroids influenced by an electric field in a microdevice. Biomed Microdevices 2018; 20:70. [PMID: 30094556 DOI: 10.1007/s10544-018-0316-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The use of electric field for cancer therapy has been proposed for a novel non-invasive cancer therapeutic approach that provides better quality of life for patients. However, argument of the efficacy hampers the therapeutic development for various cancer diseases. More scientific evidences are necessary to be addressed by basic research. The current in vitro cell culture study reports the responses of tumor spheroids after the application of an alternating electric field. Human hepatocarchinoma cells suspended in soft hydrogel were cultured in a cell culture device embedded with stimulating electrodes. Tumor spheroids gradually formed and alternating electric field was then applied during the culture course. Investigation of cell viability and cell cycle were conducted to optimize the treatment conditions. The results showed that the electric potential of 1.0 Vpp and frequency of 130 kHz was the minimal effective conditions for inhibiting tumor spheroids. Importantly, dissociation of tumor spheroids was observed after the treatment. The effectiveness of chemotherapeutic agents was shown to be enhanced while the electric filed was simultaneously applied to the tumor spheroids. These results provided solid foundation for developing the effective therapeutic strategies.
Collapse
Affiliation(s)
- Chun-Hao Huang
- PhD Program in Biomedical Engineering, College of Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Kin Fong Lei
- Graduate Institute of Medical Mechatronics, Chang Gung University, Taoyuan, Taiwan. .,Department of Mechanical Engineering, Chang Gung University, Taoyuan, Taiwan. .,Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Ngan-Ming Tsang
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan. .,Department of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
183
|
Lei KF, Hsieh SC, Kuo RL, Tsang NM. Co-Culturing Cancer Cells and Normal Cells in a Biochip under Electrical Stimulation. BIOCHIP JOURNAL 2018. [DOI: 10.1007/s13206-018-2309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
184
|
Kessler AF, Frömbling GE, Gross F, Hahn M, Dzokou W, Ernestus RI, Löhr M, Hagemann C. Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibition. Cell Death Discov 2018; 4:12. [PMID: 30210815 PMCID: PMC6125382 DOI: 10.1038/s41420-018-0079-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/22/2018] [Indexed: 01/22/2023] Open
Abstract
Tumor treating fields (TTFields) are approved for glioblastoma (GBM) therapy. TTFields disrupt cell division by inhibiting spindle fiber formation. Spindle assembly checkpoint (SAC) inhibition combined with antimitotic drugs synergistically decreases glioma cell growth in cell culture and mice. We hypothesized that SAC inhibition will increase TTFields efficacy. Human GBM cells (U-87 MG, GaMG) were treated with TTFields (200 kHz, 1.7 V/cm) and/or the SAC inhibitor MPS1-IN-3 (IN-3, 4 µM). Cells were counted after 24, 48, and 72 h of treatment and at 24 and 72 h after end of treatment (EOT). Flow cytometry, immunofluorescence microscopy, Annexin-V staining and TUNEL assay were used to detect alterations in cell cycle and apoptosis after 72 h of treatment. The TTFields/IN-3 combination decreased cell proliferation after 72 h compared to either treatment alone (-78.6% vs. TTFields, P = 0.0337; -52.6% vs. IN-3, P = 0.0205), and reduced the number of viable cells (62% less than seeded). There was a significant cell cycle shift from G1 to G2/M phase (P < 0.0001). The apoptotic rate increased to 44% (TTFields 14%, P = 0.0002; IN-3 4%, P < 0.0001). Cell growth recovered 24 h after EOT with TTFields and IN-3 alone, but the combination led to further decrease by 92% at 72 h EOT if IN-3 treatment was continued (P = 0.0288). The combination of TTFields and SAC inhibition led to earlier and prolonged effects that significantly augmented the efficacy of TTFields and highlights a potential new targeted multimodal treatment for GBM.
Collapse
Affiliation(s)
- Almuth F. Kessler
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Greta E. Frömbling
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Franziska Gross
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Mirja Hahn
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Wilfrid Dzokou
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Carsten Hagemann
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| |
Collapse
|
185
|
Benson L. Tumor Treating Fields Technology: Alternating Electric Field Therapy for the Treatment of Solid Tumors. Semin Oncol Nurs 2018; 34:137-150. [DOI: 10.1016/j.soncn.2018.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
186
|
Dahlgren PN, Bishop K, Dey S, Herbert BS, Tanaka H. Development of a New Monochrome Multiplex qPCR Method for Relative Telomere Length Measurement in Cancer. Neoplasia 2018; 20:425-431. [PMID: 29573637 PMCID: PMC5915991 DOI: 10.1016/j.neo.2018.02.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/14/2018] [Accepted: 02/19/2018] [Indexed: 01/30/2023] Open
Abstract
Excess telomere shortening has been observed in most cancer cells. The telomere quantitative polymerase chain reaction (qPCR) assay has become an important tool for epidemiological studies examining the effects of aging, stress, and other factors on the length of telomeres. Current telomere qPCR methods analyze the relative length of telomeres by amplifying telomere sequence products and normalizing with single-copy gene products. However, the current telomere qPCR does not always reflect absolute telomere length in cancer DNA. Because of genomic instability in cancer cells, we hypothesized that the use of single-copy genes (scg) is less accurate for normalizing data in cancer DNA and that new primer sets are required to better represent relative telomere length in cancer DNA. We first confirmed that cancer cells had a different copy ratio among different scg, implying that DNA is aneuploid. By using the new primer sets that amplify multiple-copy sequences (mcs) throughout the genome, the telomere qPCR results showed that the mcs primers were interchangeable with the scg primers as reference primers in normal DNA. By comparing results from the traditional southern blotting method (as kilobases) and results from monochrome multiplex qPCR using the mcs primers (as T/M ratios), we verified that the T/M ratio is highly correlated with absolute telomere length from the southern blot analysis. Together, the mcs primers were able to represent the telomere lengths accurately in cancer DNA samples. These results would allow for analyses of telomeres within cancerous DNA and the development of new, less invasive diagnostic tools for cancer.
Collapse
Affiliation(s)
- Paige N Dahlgren
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Kanokwan Bishop
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Shatovisha Dey
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Brittney-Shea Herbert
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Hiromi Tanaka
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN.
| |
Collapse
|
187
|
Stewart TS, Nagesetti A, Guduru R, Liang P, Stimphil E, Hadjikhani A, Salgueiro L, Horstmyer J, Cai R, Schally A, Khizroev S. Magnetoelectric nanoparticles for delivery of antitumor peptides into glioblastoma cells by magnetic fields. Nanomedicine (Lond) 2018; 13:423-438. [PMID: 29345190 PMCID: PMC5810849 DOI: 10.2217/nnm-2017-0300] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/11/2017] [Indexed: 12/15/2022] Open
Abstract
AIM We studied externally controlled anticancer effects of binding tumor growth inhibiting synthetic peptides to magnetoelectric nanoparticles (MENs) on treatment of glioblastomas. METHODS Hydrothermally synthesized 30-nm MENs had the core-shell composition of CoFe2O4@BaTiO3. Molecules of growth hormone-releasing hormone antagonist of the MIA class (MIA690) were chemically bound to MENs. In vitro experiments utilized human glioblastoma cells (U-87MG) and human brain microvascular endothelial cells. RESULTS The studies demonstrated externally controlled high-efficacy binding of MIA690 to MENs, targeted specificity to glioblastoma cells and on-demand release of the peptide by application of d.c. and a.c. magnetic fields, respectively. CONCLUSION The results support the use of MENs as an effective drug delivery carrier for growth hormone-releasing hormone antagonists in the treatment of human glioblastomas.
Collapse
Affiliation(s)
- Tiffanie S Stewart
- Center for Personalized Nanomedicine, Florida International University, Miami, FL, USA
- Center for Nano Science & Technology, University of Notre Dame, Notre Dame, IN, USA
| | - Abhignyan Nagesetti
- Center for Personalized Nanomedicine, Florida International University, Miami, FL, USA
| | - Rakesh Guduru
- Center for Personalized Nanomedicine, Florida International University, Miami, FL, USA
| | - Ping Liang
- Cellular Nanomed, Coral Springs, FL, USA
| | - Emmanuel Stimphil
- Center for Personalized Nanomedicine, Florida International University, Miami, FL, USA
| | - Ali Hadjikhani
- Center for Personalized Nanomedicine, Florida International University, Miami, FL, USA
| | - Luis Salgueiro
- Veterans Affairs Medical Center, University of Miami School of Medicine, Miami, FL, USA
| | | | - Renzhi Cai
- Veterans Affairs Medical Center, University of Miami School of Medicine, Miami, FL, USA
| | - Andrew Schally
- Veterans Affairs Medical Center, University of Miami School of Medicine, Miami, FL, USA
| | - Sakhrat Khizroev
- Center for Personalized Nanomedicine, Florida International University, Miami, FL, USA
- Brain Center, Miami, FL, USA
| |
Collapse
|
188
|
Tumor treating fields: a new approach to glioblastoma therapy. J Neurooncol 2018; 137:447-453. [DOI: 10.1007/s11060-018-2768-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 12/20/2022]
|
189
|
Zygogianni A, Protopapa M, Kougioumtzopoulou A, Simopoulou F, Nikoloudi S, Kouloulias V. From imaging to biology of glioblastoma: new clinical oncology perspectives to the problem of local recurrence. Clin Transl Oncol 2018; 20:989-1003. [PMID: 29335830 DOI: 10.1007/s12094-018-1831-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 01/04/2018] [Indexed: 12/13/2022]
Abstract
GBM is one of the most common and aggressive brain tumors. Surgery and adjuvant chemoradiation have succeeded in providing a survival benefit. Although most patients will eventually experience local recurrence, the means to fight recurrence are limited and prognosis remains poor. In a disease where local control remains the major challenge, few trials have addressed the efficacy of local treatments, either surgery or radiation therapy. The present article reviews recent advances in the biology, imaging and biomarker science of GBM as well as the current treatment status of GBM, providing new perspectives to the problem of local recurrence.
Collapse
Affiliation(s)
- A Zygogianni
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - M Protopapa
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - A Kougioumtzopoulou
- Radiotherapy Unit, 2nd Department of Radiology, Medical School, ATTIKON University Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462, Chaidari, Greece
| | - F Simopoulou
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - S Nikoloudi
- Radiotherapy Unit, 1st Department of Radiology, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - V Kouloulias
- Radiotherapy Unit, 2nd Department of Radiology, Medical School, ATTIKON University Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462, Chaidari, Greece.
| |
Collapse
|
190
|
Cortes AD, Seña CAD, Aggangan NS. Cytotoxic and Genotoxic Properties of a Medicinal Plant, <i>Cyperus kyllingia</i> Endl. CYTOLOGIA 2018. [DOI: 10.1508/cytologia.83.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Angelbert D. Cortes
- Biology Department, College of Natural Sciences and Mathematics, Mindanao State University Main Campus
- National Institute of Molecular Biology and Biotechnology (BIOTECH), University of the Philippines Los Baños, College
| | - Cesar A. dela Seña
- Biology Department, College of Natural Sciences and Mathematics, Mindanao State University Main Campus
| | - Nelly S. Aggangan
- National Institute of Molecular Biology and Biotechnology (BIOTECH), University of the Philippines Los Baños, College
| |
Collapse
|
191
|
Khushi M, Dean IM, Teber ET, Chircop M, Arthur JW, Flores-Rodriguez N. Automated classification and characterization of the mitotic spindle following knockdown of a mitosis-related protein. BMC Bioinformatics 2017; 18:566. [PMID: 29297284 PMCID: PMC5751558 DOI: 10.1186/s12859-017-1966-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Background Cell division (mitosis) results in the equal segregation of chromosomes between two daughter cells. The mitotic spindle plays a pivotal role in chromosome alignment and segregation during metaphase and anaphase. Structural or functional errors of this spindle can cause aneuploidy, a hallmark of many cancers. To investigate if a given protein associates with the mitotic spindle and regulates its assembly, stability, or function, fluorescence microscopy can be performed to determine if disruption of that protein induces phenotypes indicative of spindle dysfunction. Importantly, functional disruption of proteins with specific roles during mitosis can lead to cancer cell death by inducing mitotic insult. However, there is a lack of automated computational tools to detect and quantify the effects of such disruption on spindle integrity. Results We developed the image analysis software tool MatQuantify, which detects both large-scale and subtle structural changes in the spindle or DNA and can be used to statistically compare the effects of different treatments. MatQuantify can quantify various physical properties extracted from fluorescence microscopy images, such as area, lengths of various components, perimeter, eccentricity, fractal dimension, satellite objects and orientation. It can also measure textual properties including entropy, intensities and the standard deviation of intensities. Using MatQuantify, we studied the effect of knocking down the protein clathrin heavy chain (CHC) on the mitotic spindle. We analysed 217 microscopy images of untreated metaphase cells, 172 images of metaphase cells transfected with small interfering RNAs targeting the luciferase gene (as a negative control), and 230 images of metaphase cells depleted of CHC. Using the quantified data, we trained 23 supervised machine learning classification algorithms. The Support Vector Machine learning algorithm was the most accurate method (accuracy: 85.1%; area under the curve: 0.92) for classifying a spindle image. The Kruskal-Wallis and Tukey-Kramer tests demonstrated that solidity, compactness, eccentricity, extent, mean intensity and number of satellite objects (multipolar spindles) significantly differed between CHC-depleted cells and untreated/luciferase-knockdown cells. Conclusion MatQuantify enables automated quantitative analysis of images of mitotic spindles. Using this tool, researchers can unambiguously test if disruption of a protein-of-interest changes metaphase spindle maintenance and thereby affects mitosis.
Collapse
Affiliation(s)
- Matloob Khushi
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia. .,Current address: School of IT, The University of Sydney, Darlington, NSW, Australia.
| | - Imraan M Dean
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Erdahl T Teber
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Megan Chircop
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Jonathan W Arthur
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Neftali Flores-Rodriguez
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia.,Current address: School of Biomedical Sciences, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
192
|
Stupp R, Taillibert S, Kanner A, Read W, Steinberg DM, Lhermitte B, Toms S, Idbaih A, Ahluwalia MS, Fink K, Di Meco F, Lieberman F, Zhu JJ, Stragliotto G, Tran DD, Brem S, Hottinger AF, Kirson ED, Lavy-Shahaf G, Weinberg U, Kim CY, Paek SH, Nicholas G, Burna J, Hirte H, Weller M, Palti Y, Hegi ME, Ram Z. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA 2017; 318:2306-2316. [PMID: 29260225 PMCID: PMC5820703 DOI: 10.1001/jama.2017.18718] [Citation(s) in RCA: 1723] [Impact Index Per Article: 215.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Tumor-treating fields (TTFields) is an antimitotic treatment modality that interferes with glioblastoma cell division and organelle assembly by delivering low-intensity alternating electric fields to the tumor. OBJECTIVE To investigate whether TTFields improves progression-free and overall survival of patients with glioblastoma, a fatal disease that commonly recurs at the initial tumor site or in the central nervous system. DESIGN, SETTING, AND PARTICIPANTS In this randomized, open-label trial, 695 patients with glioblastoma whose tumor was resected or biopsied and had completed concomitant radiochemotherapy (median time from diagnosis to randomization, 3.8 months) were enrolled at 83 centers (July 2009-2014) and followed up through December 2016. A preliminary report from this trial was published in 2015; this report describes the final analysis. INTERVENTIONS Patients were randomized 2:1 to TTFields plus maintenance temozolomide chemotherapy (n = 466) or temozolomide alone (n = 229). The TTFields, consisting of low-intensity, 200 kHz frequency, alternating electric fields, was delivered (≥ 18 hours/d) via 4 transducer arrays on the shaved scalp and connected to a portable device. Temozolomide was administered to both groups (150-200 mg/m2) for 5 days per 28-day cycle (6-12 cycles). MAIN OUTCOMES AND MEASURES Progression-free survival (tested at α = .046). The secondary end point was overall survival (tested hierarchically at α = .048). Analyses were performed for the intent-to-treat population. Adverse events were compared by group. RESULTS Of the 695 randomized patients (median age, 56 years; IQR, 48-63; 473 men [68%]), 637 (92%) completed the trial. Median progression-free survival from randomization was 6.7 months in the TTFields-temozolomide group and 4.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.52-0.76; P < .001). Median overall survival was 20.9 months in the TTFields-temozolomide group vs 16.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.53-0.76; P < .001). Systemic adverse event frequency was 48% in the TTFields-temozolomide group and 44% in the temozolomide-alone group. Mild to moderate skin toxicity underneath the transducer arrays occurred in 52% of patients who received TTFields-temozolomide vs no patients who received temozolomide alone. CONCLUSIONS AND RELEVANCE In the final analysis of this randomized clinical trial of patients with glioblastoma who had received standard radiochemotherapy, the addition of TTFields to maintenance temozolomide chemotherapy vs maintenance temozolomide alone, resulted in statistically significant improvement in progression-free survival and overall survival. These results are consistent with the previous interim analysis. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00916409.
Collapse
Affiliation(s)
- Roger Stupp
- Lou and Jean MalnatiBrain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center of Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Sophie Taillibert
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | | | - William Read
- University of California, San Diego
- Emory University, Atlanta, Georgia
| | | | - Benoit Lhermitte
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Steven Toms
- Geisinger Health System, Danville, Pennsylvania
| | - Ahmed Idbaih
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | | | - Karen Fink
- Baylor University Medical Center, Houston, Texas
| | | | - Frank Lieberman
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jay-Jiguang Zhu
- University of Texas Health Sciences Center at Houston
- Tufts Medical Center, Boston, Massachusetts
| | | | - David D. Tran
- Washington University Barnes-Jewish Hospital, St Louis, Missouri
| | - Steven Brem
- Moffitt Cancer Center, Tampa, Florida
- University of Pennsylvania, Philadelphia
| | - Andreas F. Hottinger
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | | | | | - Chae-Yong Kim
- Seoul National University Bundang Hospital, Seoul National University College of Medicine, Bundang, Korea
| | | | - Garth Nicholas
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jordi Burna
- Hospital Universitario de Bellvitge, Barcelona, Spain
| | - Hal Hirte
- Juravinski Cancer Centre, Hamilton, Ontario, Canada
| | - Michael Weller
- University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | | | - Monika E. Hegi
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Zvi Ram
- Tel Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
193
|
Saria MG, Kesari S. Efficacy and Safety of Treating Glioblastoma With Tumor-Treating Fields Therapy. Clin J Oncol Nurs 2017; 20:S9-S13. [PMID: 27668388 DOI: 10.1188/16.cjon.s1.9-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive astrocytoma with a dismal prognosis. Since 1976, only three chemotherapeutic agents have been approved for the treatment of GBM. Tumor-treating fields (TTFields) therapy, delivered via a noninvasive device, is a new therapy approved for use in patients with recurrent GBM and in combination with temozolomide for the treatment of newly diagnosed GBM. OBJECTIVES This article reviews the mechanism of action and findings from preclinical and clinical studies supporting the use of TTFields for patients with newly diagnosed and recurrent GBM. METHODS This article provides an overview of published literature on the efficacy and safety of treating GBM with TTFields. FINDINGS For the first time in more than a decade, patients with GBM have a noninvasive treatment option that has been shown to increase progression-free survival and overall survival with minimal adverse events.
Collapse
Affiliation(s)
- Marlon G Saria
- John Wayne Cancer Institute at Providence Saint John's Health Center
| | - Santosh Kesari
- John Wayne Cancer Institute at Providence Saint John's Health Center
| |
Collapse
|
194
|
Chang E, Pohling C, Beygui N, Patel CB, Rosenberg J, Ha DH, Gambhir SS. Synergistic inhibition of glioma cell proliferation by Withaferin A and tumor treating fields. J Neurooncol 2017; 134:259-268. [PMID: 28681243 PMCID: PMC5711586 DOI: 10.1007/s11060-017-2534-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/25/2017] [Indexed: 12/18/2022]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal form of brain cancer. Standard therapies are non-specific and often of limited effectiveness; thus, efforts are underway to uncover novel, unorthodox therapies against GBM. In previous studies, we investigated Withaferin A, a steroidal lactone from Ayurvedic medicine that inhibits proliferation in cancers including GBM. Another novel approach, tumor treating fields (TTFields), is thought to disrupt mitotic spindle formation and stymie proliferation of actively dividing cells. We hypothesized that combining TTFields with Withaferin A would synergistically inhibit proliferation in glioblastoma. Human glioblastoma cells (GBM2, GBM39, U87-MG) and human breast adenocarcinoma cells (MDA-MB-231) were isolated from primary tumors. The glioma cell lines were genetically engineered to express firefly luciferase. Proliferative potential was assessed either by bioluminescence imaging or cell counting via hemocytometer. TTFields (4 V/cm) significantly inhibited growth of the four cancer cell lines tested (n = 3 experiments per time point, four measurements per sample, p < 0.02 at least; 2-way ANOVA, control vs. treatment). The combination of Withaferin A (10-100 nM) with TTFields significantly inhibited the growth of the glioma cells to a degree beyond that of Withaferin A or TTFields alone. The interaction of the Withaferin A and TTFields on glioma cells was found to be synergistic in nature (p < 0.01, n = 3 experiments). These findings were validated by both bioluminescence and hemocytometric measurements. The combination of Withaferin A with TTFields represents a novel approach to treat GBM in a manner that is likely better than either treatment alone and that is synergistic.
Collapse
Affiliation(s)
- Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Early Cancer Detection, Stanford University, Palo Alto, CA, USA
| | - Christoph Pohling
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Early Cancer Detection, Stanford University, Palo Alto, CA, USA
| | | | - Chirag B Patel
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Early Cancer Detection, Stanford University, Palo Alto, CA, USA
| | - Jarrett Rosenberg
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Early Cancer Detection, Stanford University, Palo Alto, CA, USA
| | - Dong Ho Ha
- Department of Radiology, Dong-A University Medical Center, Busan, Korea
| | - Sanjiv S Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Early Cancer Detection, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
195
|
Zhu JJ, Demireva P, Kanner AA, Pannullo S, Mehdorn M, Avgeropoulos N, Salmaggi A, Silvani A, Goldlust S, David C, Benouaich-Amiel A. Health-related quality of life, cognitive screening, and functional status in a randomized phase III trial (EF-14) of tumor treating fields with temozolomide compared to temozolomide alone in newly diagnosed glioblastoma. J Neurooncol 2017; 135:545-552. [PMID: 28849310 PMCID: PMC5700237 DOI: 10.1007/s11060-017-2601-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/19/2017] [Indexed: 12/22/2022]
Abstract
We characterized health-related quality of life (HRQoL), cognitive, and functional status in newly diagnosed glioblastoma (GBM) patients receiving Tumor treating fields (TTFields) with temozolomide (TMZ) versus TMZ alone in a planned interim analysis of a randomized phase III trial [NCT00916409], which showed significant improvement in progression-free and overall survival with TTFields/TMZ. After radiotherapy with concomitant TMZ, newly diagnosed GBM patients were randomized (2:1) to TTFields/TMZ (n = 210) or TMZ (n = 105). Interim analysis was performed in 315 patients with ≥18 months of follow-up. HRQoL, a secondary endpoint, was evaluated in per-protocol patient population and expressed as change from baseline (CFB) at 3, 6, and 9 months for each subscale in the EORTC QLQ-C30/BN20. Karnofsky performance scores (KPS) and Mini-Mental State Examination scores (MMSE) were assessed. CFB in HRQoL was balanced in treatment groups at the 12-month time point. Initially, HRQoL improved in patients treated with TTFields/TMZ (CFB3: 24% and CFB6: 13%) versus TMZ (CFB3: −7% and CFB6: −17%), though this difference was no longer evident at the 9-month point. General scales, including physical and social functioning, showed no difference at 9 and 12 months. TTFields/TMZ group reported higher concerns of “itchy skin”. KPS over 12 months was just below 90 in both groups. Cognitive status (MMSE) was stable over time. HRQoL, KPS, and MMSE were balanced in both groups over time. There was no preliminary evidence that HRQoL, cognitive, and functional status is adversely affected by the continuous use of TTFields.
Collapse
Affiliation(s)
- Jay-Jiguang Zhu
- McGovern Medical School and Memorial Hermann Hospital at Texas Medical Center, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | | | - Andrew A Kanner
- Rabin Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Susan Pannullo
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Hottinger AF, Pacheco P, Stupp R. Tumor treating fields: a novel treatment modality and its use in brain tumors. Neuro Oncol 2017; 18:1338-49. [PMID: 27664860 PMCID: PMC5035531 DOI: 10.1093/neuonc/now182] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 07/11/2016] [Indexed: 01/02/2023] Open
Abstract
Tumor treating fields (TTFields) are low-intensity electric fields alternating at an intermediate frequency (200kHz), which have been demonstrated to block cell division and interfere with organelle assembly. This novel treatment modality has shown promise in a variety of tumor types. It has been evaluated in randomized phase 3 trials in glioblastoma (GBM) and demonstrated to prolong progression-free survival (PFS) and overall survival (OS) when administered together with standard maintenance temozolomide (TMZ) chemotherapy in patients with newly diagnosed GBM. TTFields are continuously delivered by 4 transducer arrays consisting each of 9 insulated electrodes that are placed on the patient's shaved scalp and connected to a portable device. Here we summarize the preclinical data and mechanism of action, the available clinical data, and further outlook of this treatment modality in brain tumors and other cancer indications.
Collapse
Affiliation(s)
- Andreas F Hottinger
- CHUV University Hospital and Lausanne University, Departments of Clinical Neurosciences and Oncology, Lausanne, Switzerland (A.F.H., P.P.); University Hospital Zurich and University of Zurich, Department of Oncology, Zurich, Switzerland (R.S.)
| | - Patricia Pacheco
- CHUV University Hospital and Lausanne University, Departments of Clinical Neurosciences and Oncology, Lausanne, Switzerland (A.F.H., P.P.); University Hospital Zurich and University of Zurich, Department of Oncology, Zurich, Switzerland (R.S.)
| | - Roger Stupp
- CHUV University Hospital and Lausanne University, Departments of Clinical Neurosciences and Oncology, Lausanne, Switzerland (A.F.H., P.P.); University Hospital Zurich and University of Zurich, Department of Oncology, Zurich, Switzerland (R.S.)
| |
Collapse
|
197
|
Mun EJ, Babiker HM, Weinberg U, Kirson ED, Von Hoff DD. Tumor-Treating Fields: A Fourth Modality in Cancer Treatment. Clin Cancer Res 2017; 24:266-275. [PMID: 28765323 DOI: 10.1158/1078-0432.ccr-17-1117] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/11/2017] [Accepted: 07/26/2017] [Indexed: 11/16/2022]
Abstract
Despite major advances in therapy, cancer continues to be a leading cause of mortality. In addition, toxicities of traditional therapies pose a significant challenge to tolerability and adherence. TTFields, a noninvasive anticancer treatment modality, utilizes alternating electric fields at specific frequencies and intensities to selectively disrupt mitosis in cancerous cells. TTFields target proteins crucial to the cell cycle, leading to mitotic arrest and apoptosis. TTFields also facilitate an antitumor immune response. Clinical trials of TTFields have proven safe and efficacious in patients with glioblastoma multiforme (GBM), and are FDA approved for use in newly diagnosed and recurrent GBM. Trials in other localized solid tumors are ongoing. Clin Cancer Res; 24(2); 266-75. ©2017 AACR.
Collapse
Affiliation(s)
- Elijah J Mun
- University of Washington, Department of Internal Medicine, Seattle, Washington.
| | - Hani M Babiker
- Translational Genomics Research Institute (TGen), University of Arizona Cancer Center, Tucson, Arizona
| | - Uri Weinberg
- Novocure Ltd., Topaz Building, MATAM Center, Haifa, Israel
| | - Eilon D Kirson
- Novocure Ltd., Topaz Building, MATAM Center, Haifa, Israel
| | - Daniel D Von Hoff
- Virginia G. Piper Cancer Center Clinical Trials, HonorHealth Research Institute, Scottsdale, Arizona.,Translational Genomics Research Institute (TGen), Clinical Translational Research Division, Phoenix, Arizona
| |
Collapse
|
198
|
Dong Q, Fu L, Zhao Y, Liu Y, Li Q, Qiu X, Wang E. Derlin-1 is a target to improve radiotherapy effect of esophageal squamous cell carcinoma. Oncotarget 2017; 8:55135-55146. [PMID: 28903408 PMCID: PMC5589647 DOI: 10.18632/oncotarget.19069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/18/2017] [Indexed: 01/12/2023] Open
Abstract
Radiotherapy is widely used for treatment of esophageal squamous cell carcinoma (ESCC). This study aimed to explore the role of Derlin-1 on the sensitivity of ESCC to radiotherapy and its underlying mechanism. We examined the clinical significance of Derlin-1 in 125 ESCC tissues. We found that Derlin-1 protein was higher in ESCC tissues than that in normal esophageal epithelial tissues. Derlin-1 overexpression was correlated with chemoradiotherapy resistance in ESCC patients and served an independent predictor for short overall survival. siRNA knockdown and plasmid transfection were carried out in ESCC cell lines. Derlin-1 depletion inhibited cell growth while its overexpression facilitated cell growth. Derlin-1 overexpression in Eca-109 cells dramatically enhanced its resistance to radiotherapy with decreased apoptosis rate. On the contrary, Derlin-1 depletion in TE-1 cell line showed the opposite effects. In addition, radioresistance conferred by Derlin-1 was attributed to its role of activating AKT/Bcl-2 signaling pathway and reducing caspase3 cleavage. Blockage of AKT signaling attenuated the role of Derlin-1 on radioresistance. Furthermore, Derlin-1 could interact with PI3K p110α in ESCC cell lines. Taken together, Our data demonstrate that Derlin-1 overexpression predicts poor prognosis and protects ESCC from irradiation induced apoptosis through PI3K/AKT/Bcl-2 signaling pathway. Derlin-1 may serve as a novel predictor for radiosentivity and a molecular target for ESCC.
Collapse
Affiliation(s)
- Qianze Dong
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Lin Fu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yue Zhao
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yang Liu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Qingchang Li
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xueshan Qiu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Enhua Wang
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
199
|
Korshoej AR, Hansen FL, Thielscher A, von Oettingen GB, Sørensen JCH. Impact of tumor position, conductivity distribution and tissue homogeneity on the distribution of tumor treating fields in a human brain: A computer modeling study. PLoS One 2017; 12:e0179214. [PMID: 28604803 PMCID: PMC5467909 DOI: 10.1371/journal.pone.0179214] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 05/25/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Tumor treating fields (TTFields) are increasingly used in the treatment of glioblastoma. TTFields inhibit cancer growth through induction of alternating electrical fields. To optimize TTFields efficacy, it is necessary to understand the factors determining the strength and distribution of TTFields. In this study, we provide simple guiding principles for clinicians to assess the distribution and the local efficacy of TTFields in various clinical scenarios. METHODS We calculated the TTFields distribution using finite element methods applied to a realistic head model. Dielectric property estimates were taken from the literature. Twentyfour tumors were virtually introduced at locations systematically varied relative to the applied field. In addition, we investigated the impact of central tumor necrosis on the induced field. RESULTS Local field "hot spots" occurred at the sulcal fundi and in deep tumors embedded in white matter. The field strength was not higher for tumors close to the active electrode. Left/right field directions were generally superior to anterior/posterior directions. Central necrosis focally enhanced the field near tumor boundaries perpendicular to the applied field and introduced significant field non-uniformity within the tumor. CONCLUSIONS The TTFields distribution is largely determined by local conductivity differences. The well conducting tumor tissue creates a preferred pathway for current flow, which increases the field intensity in the tumor boundaries and surrounding regions perpendicular to the applied field. The cerebrospinal fluid plays a significant role in shaping the current pathways and funnels currents through the ventricles and sulci towards deeper regions, which thereby experience higher fields. Clinicians may apply these principles to better understand how TTFields will affect individual patients and possibly predict where local recurrence may occur. Accurate predictions should, however, be based on patient specific models. Future work is needed to assess the robustness of the presented results towards variations in conductivity.
Collapse
Affiliation(s)
- Anders Rosendal Korshoej
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade 44, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard 100, Aarhus N, Denmark
- * E-mail:
| | - Frederik Lundgaard Hansen
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade 44, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard 100, Aarhus N, Denmark
| | - Axel Thielscher
- Danish Research Center for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Kettegaards Allé 30, DK, Hvidovre, Denmark
- Biomedical Engineering, DTU Electro, Technical University of Denmark, Ørsteds Plads, Building 349, DK, Kgs. Lyngby, Denmark
- Max Planck Institute of Biological Cybernetics, Tübingen, Germany
| | - Gorm Burckhardt von Oettingen
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade 44, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard 100, Aarhus N, Denmark
| | - Jens Christian Hedemann Sørensen
- Aarhus University Hospital, Department of Neurosurgery, Nørrebrogade 44, Aarhus C, Denmark
- Aarhus University, Department of Clinical Medicine, Palle Juul-Jensens Boulevard 100, Aarhus N, Denmark
| |
Collapse
|
200
|
Porat Y, Giladi M, Schneiderman RS, Blat R, Shteingauz A, Zeevi E, Munster M, Voloshin T, Kaynan N, Tal O, Kirson ED, Weinberg U, Palti Y. Determining the Optimal Inhibitory Frequency for Cancerous Cells Using Tumor Treating Fields (TTFields). J Vis Exp 2017. [PMID: 28518093 PMCID: PMC5607886 DOI: 10.3791/55820] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Tumor Treating Fields (TTFields) are an effective treatment modality delivered via the continuous, noninvasive application of low-intensity (1-3 V/cm), alternating electric fields in the frequency range of several hundred kHz. The study of TTFields in tissue culture is carried out using the TTFields in vitro application system, which allows for the application of electric fields of varying frequencies and intensities to ceramic Petri dishes with a high dielectric constant (Ɛ > 5,000). Cancerous cell lines plated on coverslips at the bottom of the ceramic Petri dishes are subjected to TTFields delivered in two orthogonal directions at various frequencies to facilitate treatment outcome tests, such as cell counts and clonogenic assays. The results presented in this report demonstrate that the optimal frequency of the TTFields with respect to both cell counts and clonogenic assays is 200 kHz for both ovarian and glioma cells.
Collapse
Affiliation(s)
- Yaara Porat
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | - Moshe Giladi
- Preclinical Research Department, Novocure Ltd., Haifa, Israel;
| | | | - Roni Blat
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | - Anna Shteingauz
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | - Einav Zeevi
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | - Mijal Munster
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | - Tali Voloshin
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | - Noa Kaynan
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | - Orna Tal
- Preclinical Research Department, Novocure Ltd., Haifa, Israel
| | | | | | | |
Collapse
|