Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Sci Rep ; 10(1): 16495, 2020 10 05.
Article En | MEDLINE | ID: mdl-33020527

Cold atmospheric plasma (CAP) technology, a relatively novel technique mainly investigated as a stand-alone cancer treatment method in vivo and in vitro, is being proposed for application in conjunction with chemotherapy. In this study, we explore whether CAP, an ionized gas produced in laboratory settings and that operates at near room temperature, can enhance Temozolomide (TMZ) cytotoxicity on a glioblastoma cell line (U87MG). Temozolomide is the first line of treatment for glioblastoma, one of the most aggressive brain tumors that remains incurable despite advancements with treatment modalities. The cellular response to a single CAP treatment followed by three treatments with TMZ was monitored with a cell viability assay. According to the cell viability results, CAP treatment successfully augmented the effect of a cytotoxic TMZ dose (50 µM) and further restored the effect of a non-cytotoxic TMZ dose (10 µM). Application of CAP in conjunction TMZ increased DNA damage measured by the phosphorylation of H2AX and induced G2/M cell cycle arrest. These findings were supported by additional data indicating reduced cell migration and increased αvß3 and αvß5 cell surface integrin expression as a result of combined CAP-TMZ treatment. The data presented in this study serve as evidence that CAP technology can be a suitable candidate for combination therapy with existing chemotherapeutic drugs. CAP can also be investigated in future studies for sensitizing glioblastoma cells to TMZ and other drugs available in the market.


Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Plasma Gases/pharmacology , Temozolomide/pharmacology , Apoptosis/drug effects , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy/methods , DNA Damage/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Glioblastoma/metabolism , Humans , Integrin alphaVbeta3/metabolism , M Phase Cell Cycle Checkpoints/drug effects , Phosphorylation/drug effects , Receptors, Vitronectin/metabolism
2.
ACS Appl Mater Interfaces ; 11(34): 30621-30630, 2019 Aug 28.
Article En | MEDLINE | ID: mdl-31374163

The applications of the cold atmospheric plasma jet (CAPJ) in cancer treatment have been investigated for over a decade, focused on the effect that the CAPJ creates on cancer cells. Here we report for the first time on the impact that cells have on the CAPJ during treatment. To better understand these CAPJ-cell interactions, we analyzed the CAPJ behaviors in the presence of several normal and cancer cell lines and investigated the CAPJ selectivity. A more in-depth study of plasma self-organization patterns utilizing a model which contains a combination of normal and cancer cells reveals that the cells' capacitance can be an important predictor of plasma jet behavior. Cancer cells can direct the jet either toward or away from normal cells, which depends on the boundary condition behind the cell colony. Both experimental and theoretical results show that a grounded copper board beneath the cell-culture dish leads to opposite CPAJ behaviors compared with a floating boundary condition. In conclusion, our findings indicate that plasma can be self-adaptive toward cancer cells, and such a feature can be manipulated. Therefore, using the permittivity difference among cell lines may help us focus plasmas upon cancer cells at the vicinity of normal tissues and maximize the selectivity of plasma treatments.


Models, Biological , Neoplasms/drug therapy , Plasma Gases/pharmacology , Animals , Cell Line, Tumor , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Plasma Gases/chemistry
3.
ACS Appl Mater Interfaces ; 10(11): 9269-9279, 2018 Mar 21.
Article En | MEDLINE | ID: mdl-29473408

Cold atmospheric plasma (CAP), an ionized gas operated at near-ambient temperatures, has been introduced as a new therapeutic opportunity for treating cancers. The effectiveness of the therapy has been linked to CAP-generated reactive oxygen and nitrogen species such as hydrogen peroxide and nitrite. In this study, we monitor in real-time cancer cell response to CAP over the course of 48 h. The results demonstrate a correlation between cell viability, exposure time (30, 60, 90, and 180 s), and discharge voltage (3.16 and 3.71 kV), while stressing the likely therapeutic role of plasma-generated reactive species. A 30-60 s increase in CAP exposure time and/or a discharge voltage adjustment from 3.16 to 3.71 kV is consistently accompanied by a significant reduction in cell viability. Comparably, levels of hydrogen peroxide and nitrite vary as a function of voltage with elevated levels detected at the highest tested voltage condition of 3.71 kV. CAP ultimately initiates a reduction in cell viability and triggers apoptosis via damage to the mitochondrial membrane, while also deregulating protein synthesis. The findings presented in this study are discussed in the context of facilitating the development of an adaptive CAP platform which could improve treatment outcomes.


Cold Temperature , Apoptosis , Cell Survival , Humans , Hydrogen Peroxide , Neoplasms , Plasma Gases , Reactive Oxygen Species
4.
Cancers (Basel) ; 9(6)2017 May 30.
Article En | MEDLINE | ID: mdl-28555065

Cold atmospheric plasma (CAP) treatment is a rapidly expanding and emerging technology for cancer treatment. Direct CAP jet irradiation is limited to the skin and it can also be invoked as a supplement therapy during surgery as it only causes cell death in the upper three to five cell layers. However, the current cannulas from which the plasma emanates are too large for intracranial applications. To enhance efficiency and expand the applicability of the CAP method for brain tumors and reduce the gas flow rate and size of the plasma jet, a novel micro-sized CAP device (µCAP) was developed and employed to target glioblastoma tumors in the murine brain. Various plasma diagnostic techniques were applied to evaluate the physics of helium µCAP such as electron density, discharge voltage, and optical emission spectroscopy (OES). The direct and indirect effects of µCAP on glioblastoma (U87MG-RedFluc) cancer cells were investigated in vitro. The results indicate that µCAP generates short- and long-lived species and radicals (i.e., hydroxyl radical (OH), hydrogen peroxide (H2O2), and nitrite (NO2-), etc.) with increasing tumor cell death in a dose-dependent manner. Translation of these findings to an in vivo setting demonstrates that intracranial µCAP is effective at preventing glioblastoma tumor growth in the mouse brain. The µCAP device can be safely used in mice, resulting in suppression of tumor growth. These initial observations establish the µCAP device as a potentially useful ablative therapy tool in the treatment of glioblastoma.

5.
Biointerphases ; 11(3): 031010, 2016 09 07.
Article En | MEDLINE | ID: mdl-27604078

Nonthermal atmospheric plasma (NTAP) can be applied to living tissues and cells as a novel technology for cancer therapy. The authors report on a NTAP argon solution generated in deionized (DI) water for treating human gastric cancer cells (NCI-N87). Our findings show that the plasma generated in DI water with 30-min duration has the strongest effect on apoptosis in precultured human gastric cancer cells. This result can be attributed to the presence of reactive oxygen species (ROS) and reactive nitrogen species (RNS) produced in water during treatment. Furthermore, the data show that the elevated levels of RNS may play a more significant role than ROS in the rate of cell death.


Antineoplastic Agents/pharmacology , Argon/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Plasma Gases , Water/pharmacology , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Humans , Reactive Nitrogen Species/pharmacology , Reactive Oxygen Species/pharmacology , Time Factors
...