Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cancer Immunol Immunother ; 69(7): 1191-1204, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32144446

ABSTRACT

Tumor-treating fields (TTFields) are alternating electric fields in a specific frequency range (100-300 kHz) delivered to the human body through transducer arrays. In this study, we evaluated whether TTFields-mediated cell death can elicit antitumoral immunity and hence would be effectively combined with anti-PD-1 therapy. We demonstrate that in TTFields-treated cancer cells, damage-associated molecular patterns including high-mobility group B1 and adenosine triphosphate are released and calreticulin is exposed on the cell surface. Moreover, we show that TTFields treatment promotes the engulfment of cancer cells by dendritic cells (DCs) and DCs maturation in vitro, as well as recruitment of immune cells in vivo. Additionally, our study demonstrates that the combination of TTFields with anti-PD-1 therapy results in a significant decline of tumor volume and increase in the percentage of tumor-infiltrating leukocytes in two tumor models. In orthotopic lung tumors, these infiltrating leukocytes, specifically macrophages and DCs, showed elevated expression of PD-L1. Compatibly, cytotoxic T-cells isolated from these tumors demonstrated increased production of IFN-γ. In colon cancer tumors, T-cells infiltration was significantly increased following long treatment duration with TTFields plus anti-PD-1. Collectively, our results suggest that TTFields therapy can induce anticancer immune response. Furthermore, we demonstrate robust efficacy of concomitant application of TTFields and anti-PD-1 therapy. These data suggest that integrating TTFields with anti-PD-1 therapy may further enhance antitumor immunity, hence achieve better tumor control.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Hepatocellular/therapy , Carcinoma, Lewis Lung/therapy , Electric Stimulation Therapy/methods , Immunogenic Cell Death , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Proliferation , Combined Modality Therapy , Female , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Clin Med Insights Oncol ; 13: 1179554918825449, 2019.
Article in English | MEDLINE | ID: mdl-30728735

ABSTRACT

BACKGROUND: Tumor treating fields (TTFields) are a non-invasive antimitotic therapy that delivers alternating electric fields via the Optune® system. The Phase III EF-14 trial in newly diagnosed glioblastoma multiforme (GBM) showed significantly improved progression-free, overall and long-term survival when Optune was used together with maintenance temozolomide (TMZ) compared with TMZ alone. Compliance (average monthly use) was associated with better clinical outcome. The first-generation Optune system weighed approximately 6 pounds (~2.7 kg). The second-generation redesigned Optune system weighs 2.7 pounds (~1.2 kg). We tested and compared GBM patient experience with the second-generation system versus the first-generation system. METHODS: Ten newly diagnosed and recurrent GBM patients in Germany (median age: 52.9 years [31-79]) were prospectively monitored over the first month of transitioning from the first-generation to the second-generation Optune system. Questionnaires using a numerical analog scale assessed feedback at baseline (first generation) and after 1 month of second-generation use. RESULTS: After transitioning to the second-generation system, compliance improved by more than 10% in four patients, was maintained in five patients and decreased by more than 10% in one patient. Following transition, eight out of nine patients reported a reduction in the triggering of malfunction alarms. Self-reported patient feedback showed improved handling and portability (weight, mobility) of the second- versus the first-generation Optune system. CONCLUSIONS: This patient user survey suggests that patient satisfaction with the second-generation Optune system is improved versus the first-generation system. Improved features of the new system help patients achieve and maintain a higher rate of treatment compliance.

3.
Cell Death Dis ; 9(11): 1074, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341282

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.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Brain Neoplasms/pathology , Electric Stimulation/methods , Glioblastoma/pathology , Up-Regulation , Adenosine Triphosphate/metabolism , Aneuploidy , Animals , Autophagosomes/metabolism , Autophagy-Related Protein 7/antagonists & inhibitors , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Survival , Electric Stimulation Therapy , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Glioblastoma/therapy , Heat-Shock Proteins/metabolism , Humans , Lysosomes/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Mitosis , Rats , Vascular Endothelial Growth Factor A
4.
JAMA Oncol ; 4(4): 495-504, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29392280

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.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Physical Therapy Modalities , Quality of Life , Transcranial Direct Current Stimulation , Adult , Aged , Aged, 80 and over , Brain Neoplasms/epidemiology , Brain Neoplasms/psychology , Chemoradiotherapy/adverse effects , Chemoradiotherapy/methods , Combined Modality Therapy , Cytoreduction Surgical Procedures/adverse effects , Cytoreduction Surgical Procedures/methods , Disease-Free Survival , Female , Follow-Up Studies , Glioblastoma/epidemiology , Glioblastoma/psychology , Health Status , Humans , Male , Middle Aged , Neurosurgical Procedures/adverse effects , Neurosurgical Procedures/methods , Physical Therapy Modalities/adverse effects , Surveys and Questionnaires , Temozolomide/therapeutic use , Transcranial Direct Current Stimulation/adverse effects , Transcranial Direct Current Stimulation/methods , Treatment Outcome , Young Adult
5.
Clin Cancer Res ; 24(2): 266-275, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28765323

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.


Subject(s)
Neoplasms/therapy , Animals , Clinical Trials as Topic , Combined Modality Therapy/adverse effects , Combined Modality Therapy/instrumentation , Combined Modality Therapy/methods , Humans , Neoplasms/mortality , Neoplasms/pathology , Treatment Outcome
6.
Radiat Oncol ; 12(1): 206, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29284495

ABSTRACT

BACKGROUND: Tumor Treating Fields (TTFields) are an anti-neoplastic treatment modality delivered via application of alternating electric fields using insulated transducer arrays placed directly on the skin in the region surrounding the tumor. A Phase 3 clinical trial has demonstrated the effectiveness of continuous TTFields application in patients with glioblastoma during maintenance treatment with Temozolomide. The goal of this study was to evaluate the efficacy of combining TTFields with radiation treatment (RT) in glioma cells. We also examined the effect of TTFields transducer arrays on RT distribution in a phantom model and the impact on rat skin toxicity. METHODS: The efficacy of TTFields application after induction of DNA damage by RT or bleomycin was tested in U-118 MG and LN-18 glioma cells. The alkaline comet assay was used to measure repair of DNA lesions. Repair of DNA double strand breaks (DSBs) were assessed by analyzing γH2AX or Rad51 foci. DNA damage and repair signaled by the activation pattern of phospho-ATM (pS1981) and phospho-DNA-PKcs (pS2056) was evaluated by immunoblotting. The absorption of the RT energy by transducer arrays was measured by applying RT through arrays placed on a solid-state phantom. Skin toxicities were tested in rats irradiated daily through the arrays with 2Gy (total dose of 20Gy). RESULTS: TTFields synergistically enhanced the efficacy of RT in glioma cells. Application of TTFields to irradiated cells impaired repair of irradiation- or chemically-induced DNA damage, possibly by blocking homologous recombination repair. Transducer arrays presence caused a minor reduction in RT intensity at 20 mm and 60 mm below the arrays, but led to a significant increase in RT dosage at the phantom surface jeopardizing the "skin sparing effect". Nevertheless, transducer arrays placed on the rat skin during RT did not lead to additional skin reactions. CONCLUSIONS: Administration of TTFields after RT increases glioma cells treatment efficacy possibly by inhibition of DNA damage repair. These preclinical results support the application of TTFields therapy immediately after RT as a viable regimen to enhance RT outcome. Phantom measurements and animal models imply that it may be possible to leave the transducer arrays in place during RT without increasing skin toxicities.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Electric Stimulation Therapy , Glioma/radiotherapy , Phantoms, Imaging , Skin Diseases/prevention & control , Animals , Glioma/genetics , Glioma/pathology , Humans , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
7.
JAMA ; 318(23): 2306-2316, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29260225

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.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Electric Stimulation Therapy , Glioblastoma/drug therapy , Adult , Aged , Antineoplastic Agents, Alkylating/adverse effects , Chemoradiotherapy , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Disease-Free Survival , Female , Follow-Up Studies , Glioblastoma/radiotherapy , Glioblastoma/surgery , Humans , Maintenance Chemotherapy , Male , Middle Aged , Mitosis , Survival Analysis , Temozolomide
8.
J Vis Exp ; (123)2017 05 04.
Article in English | MEDLINE | ID: mdl-28518093

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.


Subject(s)
Colony-Forming Units Assay/methods , Electric Stimulation Therapy , Electricity , Glioma/therapy , Ovarian Neoplasms/therapy , Antineoplastic Protocols , Cell Line, Tumor , Female , Humans , Treatment Outcome
9.
Int J Cancer ; 139(12): 2850-2858, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27561100

ABSTRACT

Long-term survival rates for advanced ovarian cancer patients have not changed appreciably over the past four decades; therefore, development of new, effective treatment modalities remains a high priority. Tumor Treating Fields (TTFields), a clinically active anticancer modality utilize low-intensity, intermediate frequency, alternating electric fields. The goal of this study was to evaluate the efficacy of combining TTFields with paclitaxel against ovarian cancer cells in vitro and in vivo. In vitro application of TTFields on human ovarian cancer cell lines led to a significant reduction in cell counts as compared to untreated cells. The effect was found to be frequency and intensity dependent. Further reduction in the number of viable cells was achieved when TTFields treatment was combined with paclitaxel. The in vivo effect of the combined treatment was tested in mice orthotopically implanted with MOSE-LTICv cells. In this model, combined treatment led to a significant reduction in tumor luminescence and in tumor weight as compared to untreated mice. The feasibility of effective local delivery of TTFields to the human abdomen was examined using finite element mesh simulations performed using the Sim4life software. These simulations demonstrated that electric fields intensities inside and in the vicinity of the ovaries of a realistic human computational phantom are about 1 and 2 V/cm pk-pk, respectively, which is within the range of intensities required for TTFields effect. These results suggest that prospective clinical investigation of the combination of TTFields and paclitaxel is warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Mice , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/therapy , Treatment Outcome , Xenograft Model Antitumor Assays
10.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 6461-6464, 2016 Aug.
Article in English | MEDLINE | ID: mdl-28269726

ABSTRACT

This paper reviews the state-of-the-art in simulation-based studies of Tumor Treating Fields (TTFields) and highlights major aspects of TTFields in which simulation-based studies could affect clinical outcomes. A major challenge is how to simulate multiple scenarios rapidly for TTFields delivery. Overcoming this challenge will enable a better understanding of how TTFields distribution is correlated with disease progression, leading to better transducer array designs and field optimization procedures, ultimately improving patient outcomes.


Subject(s)
Computer Simulation , Electric Stimulation Therapy/instrumentation , Electric Stimulation Therapy/methods , Phantoms, Imaging , Animals , Cell Line, Tumor , Head , Humans , Neoplasms/therapy , Transducers
11.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 2357-2360, 2016 Aug.
Article in English | MEDLINE | ID: mdl-28268799

ABSTRACT

Tumor Treating Fields (TTFields) are low intensity alternating electric fields in the 100-500 KHz frequency range that are known to have an anti-mitotic effect on cancerous cells. In the USA, TTFields are approved by the Food and Drug Administration (FDA) for the treatment of glioblastoma (GBM) in both the newly diagnosed and recurrent settings. Optimizing treatment with TTFields requires a deep understanding of how TTFields distribute within the brain. To address this issue, simulations using realistic head models have been performed. However, the preparation of such models is time-consuming and requires a high level of expertise, limiting the usefulness of these models for systematic studies in which the testing of multiple cases is required. Here we present a platform for rapidly simulating TTFields distributions in multiple scenarios. This platform enables high throughput computational simulations to be performed, allowing comparison of field distributions within the head in multiple clinically relevant scenarios. The simulation setup is simple and intuitive, allowing non-expert users to run simulations and evaluate results, thereby providing a valuable tool for studying how to optimize TTFields delivery in the clinic.


Subject(s)
Brain Neoplasms/therapy , Electric Stimulation Therapy , Glioblastoma/therapy , Brain , Computer Simulation , Electricity , Head , Humans , Models, Theoretical
12.
Sci Rep ; 5: 18046, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26658786

ABSTRACT

Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields. TTFields are a unique anti-mitotic treatment modality delivered in a continuous, noninvasive manner to the region of a tumor. It was previously postulated that by exerting directional forces on highly polar intracellular elements during mitosis, TTFields could disrupt the normal assembly of spindle microtubules. However there is limited evidence directly linking TTFields to an effect on microtubules. Here we report that TTFields decrease the ratio between polymerized and total tubulin, and prevent proper mitotic spindle assembly. The aberrant mitotic events induced by TTFields lead to abnormal chromosome segregation, cellular multinucleation, and caspase dependent apoptosis of daughter cells. The effect of TTFields on cell viability and clonogenic survival substantially depends upon the cell division rate. We show that by extending the duration of exposure to TTFields, slowly dividing cells can be affected to a similar extent as rapidly dividing cells.


Subject(s)
Chromosome Segregation/physiology , Mitosis/physiology , Neoplasms/pathology , Spindle Apparatus/pathology , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , Electricity , Humans , MCF-7 Cells , Microtubules/metabolism , Microtubules/pathology , Neoplasms/metabolism , Rats , Rats, Inbred F344 , Tubulin/metabolism
13.
JAMA ; 314(23): 2535-43, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26670971

ABSTRACT

IMPORTANCE: Glioblastoma is the most devastating primary malignancy of the central nervous system in adults. Most patients die within 1 to 2 years of diagnosis. Tumor-treating fields (TTFields) are a locoregionally delivered antimitotic treatment that interferes with cell division and organelle assembly. OBJECTIVE: To evaluate the efficacy and safety of TTFields used in combination with temozolomide maintenance treatment after chemoradiation therapy for patients with glioblastoma. DESIGN, SETTING, AND PARTICIPANTS: After completion of chemoradiotherapy, patients with glioblastoma were randomized (2:1) to receive maintenance treatment with either TTFields plus temozolomide (n = 466) or temozolomide alone (n = 229) (median time from diagnosis to randomization, 3.8 months in both groups). The study enrolled 695 of the planned 700 patients between July 2009 and November 2014 at 83 centers in the United States, Canada, Europe, Israel, and South Korea. The trial was terminated based on the results of this planned interim analysis. INTERVENTIONS: Treatment with TTFields was delivered continuously (>18 hours/day) via 4 transducer arrays placed on the shaved scalp and connected to a portable medical device. Temozolomide (150-200 mg/m2/d) was given for 5 days of each 28-day cycle. MAIN OUTCOMES AND MEASURES: The primary end point was progression-free survival in the intent-to-treat population (significance threshold of .01) with overall survival in the per-protocol population (n = 280) as a powered secondary end point (significance threshold of .006). This prespecified interim analysis was to be conducted on the first 315 patients after at least 18 months of follow-up. RESULTS: The interim analysis included 210 patients randomized to TTFields plus temozolomide and 105 randomized to temozolomide alone, and was conducted at a median follow-up of 38 months (range, 18-60 months). Median progression-free survival in the intent-to-treat population was 7.1 months (95% CI, 5.9-8.2 months) in the TTFields plus temozolomide group and 4.0 months (95% CI, 3.3-5.2 months) in the temozolomide alone group (hazard ratio [HR], 0.62 [98.7% CI, 0.43-0.89]; P = .001). Median overall survival in the per-protocol population was 20.5 months (95% CI, 16.7-25.0 months) in the TTFields plus temozolomide group (n = 196) and 15.6 months (95% CI, 13.3-19.1 months) in the temozolomide alone group (n = 84) (HR, 0.64 [99.4% CI, 0.42-0.98]; P = .004). CONCLUSIONS AND RELEVANCE: In this interim analysis of 315 patients with glioblastoma who had completed standard chemoradiation therapy, adding TTFields to maintenance temozolomide chemotherapy significantly prolonged progression-free and overall survival. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00916409.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Electric Stimulation Therapy/methods , Glioblastoma/therapy , Maintenance Chemotherapy/methods , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Canada , Carmustine/therapeutic use , Chemoradiotherapy , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Dacarbazine/therapeutic use , Disease Progression , Disease-Free Survival , Early Termination of Clinical Trials , Electric Stimulation Therapy/adverse effects , Europe , Female , Glioblastoma/mortality , Humans , Israel , Male , Middle Aged , Republic of Korea , Temozolomide , United States , Young Adult
14.
World J Surg Oncol ; 13: 316, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26558989

ABSTRACT

BACKGROUND: Optune™, previously known as the NovoTTF-100A System™, generates Tumor Treating Fields (TTFields), an effective anti-mitotic therapy for glioblastoma. The system delivers intermediate frequency, alternating electric fields to the supratentorial brain. Patient therapy is personalized by configuring transducer array layout placement on the scalp to the tumor site using MRI measurements and the NovoTAL System. Transducer array layout mapping optimizes therapy by maximizing electric field intensity to the tumor site. This study evaluated physician performance in conducting transducer array layout mapping using the NovoTAL System compared with mapping performed by the Novocure in-house clinical team. METHODS: Fourteen physicians (7 neuro-oncologists, 4 medical oncologists, and 3 neurosurgeons) evaluated five blinded cases of recurrent glioblastoma and performed head size and tumor location measurements using a standard Digital Imaging and Communications in Medicine reader. Concordance with Novocure measurement and intra- and inter-rater reliability were assessed using relevant correlation coefficients. The study criterion for success was a concordance correlation coefficient (CCC) >0.80. RESULTS: CCC for each physician versus Novocure on 20 MRI measurements was 0.96 (standard deviation, SD ± 0.03, range 0.90-1.00), indicating very high agreement between the two groups. Intra- and inter-rater reliability correlation coefficients were similarly high: 0.83 (SD ±0.15, range 0.54-1.00) and 0.80 (SD ±0.18, range 0.48-1.00), respectively. CONCLUSIONS: This user study demonstrated an excellent level of concordance between prescribing physicians and Novocure in-house clinical teams in performing transducer array layout planning. Intra-rater reliability was very high, indicating reproducible performance. Physicians prescribing TTFields, when trained on the NovoTAL System, can independently perform transducer array layout mapping required for the initiation and maintenance of patients on TTFields therapy.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/surgery , Electric Stimulation Therapy/instrumentation , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Magnetic Resonance Imaging , Observer Variation , Reproducibility of Results , Scalp , Transducers
15.
Semin Oncol ; 41 Suppl 6: S35-41, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25213867

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related deaths worldwide. Common treatment modalities for NSCLC include surgery, radiotherapy, chemotherapy, and, in recent years, the clinical management paradigm has evolved with the advent of targeted therapies. Despite such advances, the impact of systemic therapies for advanced disease remains modest, and as such, the prognosis for patients with NSCLC remains poor. Standard modalities are not without their respective toxicities and there is a clear need to improve both efficacy and safety for current management approaches. Tumor-treating fields (TTFields) are low-intensity, intermediate-frequency alternating electric fields that disrupt proper spindle microtubule arrangement, thereby leading to mitotic arrest and ultimately to cell death. We evaluated the effects of combining TTFields with standard chemotherapeutic agents on several NSCLC cell lines, both in vitro and in vivo. Frequency titration curves demonstrated that the inhibitory effects of TTFields were maximal at 150 kHz for all NSCLC cell lines tested, and that the addition of TTFields to chemotherapy resulted in enhanced treatment efficacy across all cell lines. We investigated the response of Lewis lung carcinoma and KLN205 squamous cell carcinoma in mice treated with TTFields in combination with pemetrexed, cisplatin, or paclitaxel and compared these to the efficacy observed in mice exposed only to the single agents. Combining TTFields with these therapeutic agents enhanced treatment efficacy in comparison with the respective single agents and control groups in all animal models. Together, these findings suggest that combining TTFields therapy with chemotherapy may provide an additive efficacy benefit in the management of NSCLC.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Lewis Lung/therapy , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/therapy , Electric Stimulation Therapy , Lung Neoplasms/therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , Apoptosis , Carcinoma, Lewis Lung/mortality , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Cisplatin/administration & dosage , Combined Modality Therapy , Glutamates/administration & dosage , Guanine/administration & dosage , Guanine/analogs & derivatives , Humans , In Vitro Techniques , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Paclitaxel/administration & dosage , Pemetrexed , Survival Rate , Treatment Outcome , Tumor Cells, Cultured
16.
Pancreatology ; 14(1): 54-63, 2014.
Article in English | MEDLINE | ID: mdl-24555979

ABSTRACT

OBJECTIVES: Tumor Treating Fields (TTFields) are a non-invasive cancer treatment modality approved for the treatment of patients with recurrent glioblastoma. The present study determined the efficacy and mechanism of action of TTFields in preclinical models of pancreatic cancer. METHODS: The effect of TTFields in vitro was assessed using cell counts, clonogenic assays, cell cycle analysis and analysis of mitotic figures. The effect in vivo effect was studied in the PC1-0 hamster pancreatic cancer model. RESULTS: Application of TTFields in vitro showed a significant decrease in cell count, an increase in cell volume and reduced clonogenicity. Further analysis demonstrated significant increase in the number of abnormal mitotic figures, as well as a decrease in G2-M cell population. In hamsters with orthotopic pancreatic tumors, TTFields significantly reduced tumor volume accompanied by an increase in the frequency of abnormal mitotic events. TTFields efficacy was enhanced both in vitro and in vivo when combined with chemotherapy. CONCLUSIONS: These results provide the first evidence that TTFields serve as an effective antimitotic treatment in preclinical pancreatic cancer models and have a long term negative effect on cancer cell survival. These results make TTFields an attractive candidate for testing in the treatment of patients with pancreatic cancer.


Subject(s)
Mitosis/drug effects , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Size/drug effects , Combined Modality Therapy , Cricetinae , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Electricity , Humans , Male , Mesocricetus , Pancreatic Neoplasms/drug therapy , Treatment Outcome , Tumor Stem Cell Assay , Gemcitabine
17.
Eur J Cancer ; 48(14): 2192-202, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22608262

ABSTRACT

PURPOSE: NovoTTF-100A is a portable device delivering low-intensity, intermediate frequency electric fields via non-invasive, transducer arrays. Tumour Treatment Fields (TTF), a completely new therapeutic modality in cancer treatment, physically interfere with cell division. METHODS: Phase III trial of chemotherapy-free treatment of NovoTTF (20-24h/day) versus active chemotherapy in the treatment of patients with recurrent glioblastoma. Primary end-point was improvement of overall survival. RESULTS: Patients (median age 54 years (range 23-80), Karnofsky performance status 80% (range 50-100) were randomised to TTF alone (n=120) or active chemotherapy control (n=117). Number of prior treatments was two (range 1-6). Median survival was 6.6 versus 6.0 months (hazard ratio 0.86 [95% CI 0.66-1.12]; p=0.27), 1-year survival rate was 20% and 20%, progression-free survival rate at 6 months was 21.4% and 15.1% (p=0.13), respectively in TTF and active control patients. Responses were more common in the TTF arm (14% versus 9.6%, p=0.19). The TTF-related adverse events were mild (14%) to moderate (2%) skin rash beneath the transducer arrays. Severe adverse events occurred in 6% and 16% (p=0.022) of patients treated with TTF and chemotherapy, respectively. Quality of life analyses favoured TTF therapy in most domains. CONCLUSIONS: This is the first controlled trial evaluating an entirely novel cancer treatment modality delivering electric fields rather than chemotherapy. No improvement in overall survival was demonstrated, however efficacy and activity with this chemotherapy-free treatment device appears comparable to chemotherapy regimens that are commonly used for recurrent glioblastoma. Toxicity and quality of life clearly favoured TTF.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Electric Stimulation Therapy , Glioblastoma/therapy , Neoplasm Recurrence, Local , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Disease-Free Survival , Electric Stimulation Therapy/adverse effects , Europe , Female , Glioblastoma/drug therapy , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Israel , Kaplan-Meier Estimate , Karnofsky Performance Status , Magnetic Resonance Imaging , Male , Middle Aged , Proportional Hazards Models , Quality of Life , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , United States , Young Adult
18.
Antimicrob Agents Chemother ; 54(8): 3212-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20547811

ABSTRACT

High-frequency, low-intensity electric fields generated by insulated electrodes have previously been shown to inhibit bacterial growth in vitro. In the present study, we tested the effect of these antimicrobial fields (AMFields) on the development of lung infection caused by Pseudomonas aeruginosa in mice. We demonstrate that AMFields (10 MHz) significantly inhibit bacterial growth in vivo, both as a stand-alone treatment and in combination with ceftazidime. In addition, we show that peripheral (skin) heating of about 2 degrees C can contribute to bacterial growth inhibition in the lungs of mice. We suggest that the combination of alternating electric fields, together with the heat produced during their application, may serve as a novel antibacterial treatment modality.


Subject(s)
Electricity , Lung Diseases/therapy , Pseudomonas Infections/therapy , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/radiation effects , Animals , Anti-Bacterial Agents/therapeutic use , Ceftazidime/therapeutic use , Colony Count, Microbial , Disease Models, Animal , Electrodes , Female , Hot Temperature , Humans , Lung/microbiology , Lung/pathology , Lung Diseases/drug therapy , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Mice, Inbred ICR , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Treatment Outcome
19.
BMC Cancer ; 10: 229, 2010 May 23.
Article in English | MEDLINE | ID: mdl-20492723

ABSTRACT

BACKGROUND: Exposure of cancer cells to chemotherapeutic agents may result in reduced sensitivity to structurally unrelated agents, a phenomenon known as multidrug resistance, MDR. The purpose of this study is to investigate cell growth inhibition of wild type and the corresponding MDR cells by Tumor Treating Fields--TTFields, a new cancer treatment modality that is free of systemic toxicity. The TTFields were applied alone and in combination with paclitaxel and doxorubicin. METHODS: Three pairs of wild type/MDR cell lines, having resistivity resulting from over-expression of ABC transporters, were studied: a clonal derivative (C11) of parental Chinese hamster ovary AA8 cells and their emetine-resistant sub-line EmtR1; human breast cancer cells MCF-7 and their mitoxantrone-resistant sub lines MCF-7/Mx and human breast cancer cells MDA-MB-231 and their doxorubicin resistant MDA-MB-231/Dox cells. TTFields were applied for 72 hours with and without the chemotherapeutic agents. The numbers of viable cells in the treated cultures and the untreated control groups were determined using the XTT assay. Student t-test was applied to asses the significance of the differences between results obtained for each of the three cell pairs. RESULTS: TTFields caused a similar reduction in the number of viable cells of wild type and MDR cells. Treatments by TTFields/drug combinations resulted in a similar increased reduction in cell survival of wild type and MDR cells. TTFields had no effect on intracellular doxorubicin accumulation in both wild type and MDR cells. CONCLUSIONS: The results indicate that TTFields alone and in combination with paclitaxel and doxorubicin effectively reduce the viability of both wild type and MDR cell sub-lines and thus can potentially be used as an effective treatment of drug resistant tumors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Electric Stimulation , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Antineoplastic Agents/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Doxorubicin/metabolism , Doxorubicin/pharmacology , Emetine/pharmacology , Female , Humans , Inhibitory Concentration 50 , Mitoxantrone/pharmacology , Neoplasm Proteins/genetics , Paclitaxel/pharmacology , Time Factors , Up-Regulation
20.
Clin Exp Metastasis ; 26(7): 633-40, 2009.
Article in English | MEDLINE | ID: mdl-19387848

ABSTRACT

Tumor treating fields (TTFields) are low intensity, intermediate frequency, alternating electric fields used to treat cancerous tumors. This novel treatment modality effectively inhibits the growth of solid tumors in vivo and has shown promise in pilot clinical trials in patients with advanced stage solid tumors. TTFields were tested for their potential to inhibit metastatic spread of solid tumors to the lungs in two animal models: (1) Mice injected with malignant melanoma cells (B16F10) into the tail vein, (2) New Zealand White rabbits implanted with VX-2 tumors within the kidney capsule. Mice and rabbits were treated using two-directional TTFields at 100-200 kHz. Animals were either monitored for survival, or sacrificed for pathological and histological analysis of the lungs. The total number of lung surface metastases and the absolute weight of the lungs were both significantly lower in TTFields treated mice then in sham control mice. TTFields treated rabbits survived longer than sham control animals. This extension in survival was found to be due to an inhibition of metastatic spread, seeding or growth in the lungs of TTFields treated rabbits compared to controls. Histologically, extensive peri- and intra-tumoral immune cell infiltration was seen in TTFields treated rabbits only. These results raise the possibility that in addition to their proven inhibitory effect on the growth of solid tumors, TTFields may also have clinical benefit in the prevention of metastatic spread from primary tumors.


Subject(s)
Electricity , Lung Neoplasms/secondary , Melanoma/pathology , Neoplasm Metastasis/prevention & control , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...