scholarly journals Towards Solid Tumor Treatment by Nanosecond Pulsed Electric Fields

2009 ◽  
Vol 8 (4) ◽  
pp. 289-306 ◽  
Author(s):  
Axel T. Esser ◽  
Kyle C. Smith ◽  
T. R. Gowrishankar ◽  
James C. Weaver

Local and drug-free solid tumor ablation by large nanosecond pulsed electric fields leads to supra-electroporation of all cellular membranes and has been observed to trigger nonthermal cell death by apoptosis. To establish pore-based effects as the underlying mechanism inducing apoptosis, we use a multicellular system model (spatial scale 100 μm) that has irregularly shaped liver cells and a multiscale liver tissue model (spatial scale 200 mm). Pore histograms for the multicellular model demonstrate the presence of only nanometer-sized pores due to nanosecond electric field pulses. The number of pores in the plasma membrane is such that the average tissue conductance during nanosecond electric field pulses is even higher than for longer irreversible electroporation pulses. It is shown, however, that these nanometer-sized pores, although numerous, only significantly change the permeability of the cellular membranes to small ions, but not to larger molecules. Tumor ablation by nanosecond pulsed electric fields causes small to moderate temperature increases. Thus, the underlying mechanism(s) that trigger cell death by apoptosis must be non-thermal electrical interactions, presumably leading to different ionic and molecular transport than for much longer irreversible electroporation pulses.

Cancers ◽  
2020 ◽  
Vol 12 (5) ◽  
pp. 1132 ◽  
Author(s):  
Philip M. Graybill ◽  
Rafael V. Davalos

Pulsed electric fields (PEFs) have become clinically important through the success of Irreversible Electroporation (IRE), Electrochemotherapy (ECT), and nanosecond PEFs (nsPEFs) for the treatment of tumors. PEFs increase the permeability of cell membranes, a phenomenon known as electroporation. In addition to well-known membrane effects, PEFs can cause profound cytoskeletal disruption. In this review, we summarize the current understanding of cytoskeletal disruption after PEFs. Compiling available studies, we describe PEF-induced cytoskeletal disruption and possible mechanisms of disruption. Additionally, we consider how cytoskeletal alterations contribute to cell–cell and cell–substrate disruption. We conclude with a discussion of cytoskeletal disruption-induced anti-vascular effects of PEFs and consider how a better understanding of cytoskeletal disruption after PEFs may lead to more effective therapies.


2019 ◽  
Vol 2019 ◽  
pp. 1-10
Author(s):  
Yan Mi ◽  
Quan Liu ◽  
Pan Li ◽  
Jin Xu

Carbon nanotubes (CNTs) with large aspect ratios and excellent electrical properties can enhance the killing effect of nanosecond pulsed electric fields (nsPEFs) on tumor cells, which can improve the electrical safety of nsPEF during tumor treatment. To study the mechanism of the CNT-enhanced killing effect of a nsPEF on tumor cells, a spherical, single-cell, five-layer dielectric model containing randomly distributed CNTs was established using COMSOL and MATLAB, and then, the effects of the addition of CNTs on the electric field and the electroporation effect on the inner and outer membranes were analyzed. The results showed that CNTs can enhance the local electric field strength due to a lightning rod effect, and the closer the CNT tip was to the cell, the greater the electric field strength was around the cell. This increase in the local electric field strength near the cells enhanced the electroporation effects, including pore density, pore area, and pore flux. The simulation results presented in this paper provide theoretical guidance for subsequent development of nsPEF combined with CNTs for use in both cell and tissue experiments.


2018 ◽  
Vol 101 (3) ◽  
pp. 38-48 ◽  
Author(s):  
RYUYA ADACHI ◽  
SHOTA HATAYAMA ◽  
NOBUAKI OHNISHI ◽  
SUNAO KATSUKI ◽  
TOSHIAKI WADA ◽  
...  

2017 ◽  
Vol 137 (6) ◽  
pp. 320-327
Author(s):  
Ryuta Andachi ◽  
Shota Hatayama ◽  
Nobuaki Ohnishi ◽  
Sunao Katsuki ◽  
Toshiaki Wada ◽  
...  

Cancers ◽  
2019 ◽  
Vol 11 (12) ◽  
pp. 2034 ◽  
Author(s):  
Alessandra Rossi ◽  
Olga N. Pakhomova ◽  
Peter A. Mollica ◽  
Maura Casciola ◽  
Uma Mangalanathan ◽  
...  

Depending on the initiating stimulus, cancer cell death can be immunogenic or non-immunogenic. Inducers of immunogenic cell death (ICD) rely on endoplasmic reticulum (ER) stress for the trafficking of danger signals such as calreticulin (CRT) and ATP. We found that nanosecond pulsed electric fields (nsPEF), an emerging new modality for tumor ablation, cause the activation of the ER-resident stress sensor PERK in both CT-26 colon carcinoma and EL-4 lymphoma cells. PERK activation correlates with sustained CRT exposure on the cell plasma membrane and apoptosis induction in both nsPEF-treated cell lines. Our results show that, in CT-26 cells, the activity of caspase-3/7 was increased fourteen-fold as compared with four-fold in EL-4 cells. Moreover, while nsPEF treatments induced the release of the ICD hallmark HMGB1 in both cell lines, extracellular ATP was detected only in CT-26. Finally, in vaccination assays, CT-26 cells treated with nsPEF or doxorubicin equally impaired the growth of tumors at challenge sites eliciting a protective anticancer immune response in 78% and 80% of the animals, respectively. As compared to CT-26, both nsPEF- and mitoxantrone-treated EL-4 cells had a less pronounced effect and protected 50% and 20% of the animals, respectively. These results support our conclusion that nsPEF induce ER stress, accompanied by bona fide ICD.


Sign in / Sign up

Export Citation Format

Share Document