Application of Cold Atmospheric Plasma (CAP) in Cancer Therapy: A Review

authors:

avatar Reza Reiazi 1 , * , avatar Mohammad Esmaeil Akbari 2 , avatar Amir Norozi 1 , avatar Maryam Etedadialiabadi 3

Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, IR Iran
Shadi Beheshty University of Medical Sciences, School of Medicine, Tehran, IR Iran
Milad Hospital, Radiology Department, Tehran, IR Iran

how to cite: Reiazi R, Akbari M E, Norozi A, Etedadialiabadi M. Application of Cold Atmospheric Plasma (CAP) in Cancer Therapy: A Review. Int J Cancer Manag. 2017;10(3):e8728. https://doi.org/10.5812/ijcp.8728.

Abstract

Background:

Recently the use of cold atmospheric plasma (CAP) for the treatment of cancers called “Plasma Oncology” has gained promising results. The developments have raised the hope that this technology could be an interesting new therapeutic approach in the treatment of cancer.

Methods:

The process of this study includes a narrative review in two stages. At the first stage “literature overview”, all mechanisms of action related to cold atmospheric plasma have been reviewed. To relevant as much as papers for this review, multiple effective techniques have been applied in our search strategy. An extensive search of the published literature has been conducted.

Results:

Results of this study include three sections as follow: the mechanism of action, in vivo and in vitro findings, and studied focused on selectivity effect of CAP. According to several publications, reactive oxidative species (ROS) can be the major cause of the biological effect of plasma. Several publications concerning in vivo and in vitro studies on different cell lines and selectivity effect showed that there are promising results in favor of anti-cancer effect of CAP.

Conclusions:

The results of several studies that are summarized within this review show that CAP is effective against cancer which also indicate that CAP seems to be selective for cancer cells compared to non-neoplastic cells. It has been concluded that the feasibility of applying CAP for treating human tissue has already gained momentum although future studies require more studies for clarifying that if CAP is capable to discriminate between normal and malignant cells or not.

1. Introduction

Surgical oncology can be considered somehow as the important type of cancer therapy which is playing an important role as an effective treatment in cancer therapy (1). In Surgical oncology there is an agreement regarding Tumor R0-resection which is a total excision of a tumor with an adequate surgical margin. Moreover, the most important aspect of successful cancer therapy is the selective eradication of cancer cell without influencing the healthy tissue. This latter issue imposed complications of complete local excision of malignant cells (2-4). Therefore, challenging issues in this field are removing microscopic residues to prevent cancer-positive surgical margins, and simultaneously distance reduction between excised tumor and surrounding normal tissue. For these reasons new therapeutic applications capable of satisfying above conditions are required.

In 1879, William Crookes identified that 99% of the universe is made up of a matter other than liquid, gas, and solid, referred to as the fourth state of matter or plasma (5). Very soon after this discovery the encouraging possibilities of the application of cold plasma, better to say cold atmospheric plasma (CAP) in medicine has been claimed by most researchers. Cold atmospheric plasma (CAP) is a gas which is partially ionized and includes clouds of ions, electrons, and reactive neutral species like reactive oxygen species (ROS), hydroxyl radicals (HO), and nitrogen dioxide (NO) (6). CAP has shown significant potentials for various biomedical applications such as sterilization of infected tissues (7, 8), inactivation of microorganisms (9), wound healing (10, 11), skin regeneration (12), blood coagulation (13), tooth bleaching (14), and the last but not least in cancer therapy (15-17). CAP can directly affect malignant cells and tissue but such direct application with low plasma concentration can just be limited to the narrow depth and so can be invoked as a supplement during open surgery something like Electron Intraoperative radiotherapy (EIORT). Furthermore, a selectivity effect of CAP has been claimed by some researchers. It means cancer cells are more sensitive to its destructive effect than normal cells, which can make cold atmospheric plasma a promising application in cancer therapy. So it can be considered as a selective treatment option which just affects the cancer cells while leaving normal cells intact. This selectivity has been considered as an interesting ability of CAP in cancer surgery on which free surgical margin playing a crucial role in overall survival after surgical resection. In this study we made an investigation on anti-cancer effects of CAP through reviewing recent works in this field.

2. Methods

The process of this study includes a narrative review in two stages. At the first stage “literature overview”, all mechanism of action related to cold atmospheric plasma have been reviewed. In the second- “effectiveness review”-stage, we reviewed the outcome results regarding in vivo and in vitro studies related to anti cancer effects of CAP. To relevant as much as papers for this review, multiple effective techniques have been applied in our search strategy. An extensive search of the published literature has been conducted on the MEDLINE®, Pubmed® and EMBASE® using the following key words (Plasma OR Cold Atmospheric Plasma) AND (Cancer OR neoplasm OR carcinoma OR cancer surgery) AND (mechanism of action OR biology OR in vivo OR in vitro OR anti cancer effect OR selectivity) AND (lung carcinoma OR hepatocellular carcinoma OR neuroblastoma OR skin carcinoma OR melanoma OR colon carcinoma OR pancreatic carcinoma OR bladder carcinoma OR cervical carcinoma OR breast cancer). Also the reference lists of each article were reviewed in detail to find additional articles. Based on the above selection criteria, 75 articles were introduced to this study and finally 60 articles have been used to write this article. Most of these articles have focused on theoretical basis of our search targets. Each article was reviewed independently in full text, the relevance of retrieved articles were evaluated, and recorded the main findings of each study in a table.

3. Results

Mechanisms of action: Table 1 shows a summary of the most stated mechanism of action of CAP on different cancer cells. According to several authors, reactive oxidative species (ROS) produced by plasma can causes morphological changes, depolarization of the membrane, lipid peroxidation, and DNA damage on cells (9, 18). The anti-neoplastic effects of CAP depend primarily on the delivery of reactive oxygen and nitrogen species (ROS, RNS) (19). It has been verified that ROS can affect the proteins responsible for mitochondrial permeability which consequently will result in triggering the intrinsic apoptotic cascade (20). It has been documented that ROS are generated in cells exposed to a stress condition such as hypoxia, chemicals agents, UV radiation , etc. which cause damages to intracellular organelles and membranes, proteins, DNA, and lipids resulting in cell death in apoptotic way. Beside this, there different mechanisms of CAP in cancer cells were studied in recent papers which include: Activation of genes such as p53 protein (21) and p21 CDK inhibitor (22), Cell cycle arrest at the G2/M and S phase (23), ROS mediated cell cycle arrest (24) and Apoptosis via dysfunction of mitochondria (25). Also studies have shown that mitochondrial enzyme activity and its membrane potential followed by respiration rate are significantly decreased in cancer cells after CAP treatment (26). It has been shown that CAP is capable of controlling the concentrations of intracellular ROS, NO, and lipid peroxide (27). Also alteration of the redox state of Prxs reversibly following induction of peroxides will promote cell growth arrest resulted from CAP-driven alteration of ROS (28). Also essential cellular signaling pathways and functions of proteins may be disturbed or disrupted resulted from a strong and sustained disruption of redox signaling, as result of exposing to plasma (29). General conclusions about the anti-cancer mechanism of CAP can be summarized as below: 1- increasing intracellular ROS, which causes DNA double strands break (DSB) (19, 30). 2- Serious DNA damage which result in the cell cycle arrest (31), apoptosis or necrosis with a dose-dependent pattern (32). 3-producing H2O2 and NO which are key molecules for killing cancer cells (33). 4-stronger resistance of normal cell to CAP than cancer cells which make a killing preference accompanied with the distinct ROS levels and DSB among cancer cells and normal cells (34). Moreover, recently, CAP irradiated media has been considered as effectively as direct application of CAP treatment to kill cancer cells (35, 36).

Table 1.

A Summary of the Most Stated Mechanism of Action of CAP on Cancer Cells

AuthorMechanism of ActionCell TypeSelectivityaYearb
Lee et al. (37)CAP-generated Nitric Oxide (NO) radicalsSquamous cell carcinomaYes2016
Wang et al. (38)Alteration of BrCa cell surface receptor functionsHuman metastatic breast cancer (BrCa) cellsYes2013
Preston et al. (39)HNSCC cellsYes2014
Tuhvatulin et al. (21)Activation of p53 proteinHuman Colon Carcinoma CellsNo2012
Volotskova et al. (23)Cell cycle arrest at the G2/M and S phasePAM 212 cellsYes2012
Ahn et al. (25)Apoptosis via dysfunction of mitochondriaHeLa cancer cellsNo2011
Panngom et al. (26)Decrease of mitochondrial membrane potential, mitochondrial enzyme activity and respiration rateHuman lung cancer cellYes2013
Koritzer et al. (30)DNA double strands break resulted from ncreasing intracellular ROSChemoresistant glioma cellsYes2013

Based on the above mentioned anti-cancer effects, many studies have shown that CAP had significant success both in vitro and in vivo but mostly in the case of in vitro studies. Many cancer types have been investigated both in vitro (40, 41) and in vivo (42) studies on lung carcinoma. Also other in vitro studies on hepatocellular carcinoma (43), neuroblastoma (44), skin carcinoma (23), melanoma (45), colon carcinoma (46), pancreatic carcinoma (47), bladder carcinoma (40), cervical carcinoma (25, 48) and breast cancer (49) show the same promising result in favor of anti-cancer effects of CAP. Interesting results of anti-cancer effects of CAP have been shown in breast cancer surgery in which nonselective and incomplete tumor ablation impose severe limitations (49).

Also selectively ablation of ablate metastatic BrCa cells in vitro without damaging healthy MSCs impose new era in the application of CAP in cancer treatment. This result suggests a selective treatment of CAP in breast cancerous cells which can result in maintaining healthy cells and the tissue intact but tumor remediation accompanying near complete ablation (36). In case of normal cells, studies have shown that plasma has minimal impact on normal cellular conditions so it can leave their normal cells unaffected while selectively ablate corresponding cancer cells. For instance, media pH levels (sign for producing reactive agents) and thermal effects associated with cold plasma remain unchanged after CAP treatment (50).

A study showed an important factor that makes cancer cells more sensitive to CAP treatment is their higher percentage of cells in the S phase of the cell cycle (23). In a study by Lee et al. it was shown that CAP-generated nitric oxide (NO) radicals can selectively kill oral squamous cell carcinoma cells (37). Wang et al. showed that human metastatic breast cancer (BrCa) cells can be selectively ablated after CAP treatment using optimized plasma parameters. Also this study showed that healthy human bone marrow mesenchymal stem cells (MSCs) have not been affected (38). The same result has been shown regarding HNSCC cells applying a feasible therapeutic strategy if coupled with endoscopic technology (39). Also in this study the adjuvant application of CAP prior to surgery has been proposed.

4. Conclusions

It has been shown that recurrence-free survival in the management of operable cancers with curative approach depends primarily to surgical free margin or tumor-negative surgical margins. Moreover, this review showed that one of the promising applications of Cold atmospheric plasma (CAP) is a selective eradication of cancer cells. Based on the above mentioned characteristic of plasma, an improving tumor local control while minimizing associated side effects of surgical radicality can be considered after Local Intraoperative CAP treatment during surgery.

Till now CAP has shown a wide range of activity regarding freely suspended cells which can be further extended to systemic applications. The feasibility of applying CAP for treating human tissue has already gained momentum although future studies required for clarifying that CAP is capable to discriminate between normal and malignant cells or not.

Unfortunately this study failed to find a study focusing on investigation on anti-cancer effects of CAP on human volunteers. Also most of the articles were concentrated on positive effects of CAP. For these reasons, future in vivo and human studies will give more clear results companying an insight into probable shortcomings and disadvantages of CAP anti-cancer effects.

References

  • 1.

    Pollock RE, Morton DL. The Contemporary Role of Surgical Oncology. In: Kufe DW, Pollock RE, Weichselbaum RR, editors. Holland-Frei Cancer Medicine. Hamilton (ON): BC Decker; 2003.

  • 2.

    Sooriakumaran P, Srivastava A, Shariat SF, Stricker PD, Ahlering T, Eden CG, et al. A multinational, multi-institutional study comparing positive surgical margin rates among 22393 open, laparoscopic, and robot-assisted radical prostatectomy patients. Eur Urol. 2014;66(3):450-6. [PubMed ID: 24290695]. https://doi.org/10.1016/j.eururo.2013.11.018.

  • 3.

    Sutherland SE, Resnick MI, Maclennan GT, Goldman HB. Does the size of the surgical margin in partial nephrectomy for renal cell cancer really matter? J Urol. 2002;167(1):61-4. [PubMed ID: 11743276].

  • 4.

    Yafi FA, Aprikian AG, Fradet Y, Chin JL, Izawa J, Rendon R, et al. Surveillance guidelines based on recurrence patterns after radical cystectomy for bladder cancer: the Canadian Bladder Cancer Network experience. BJU Int. 2012;110(9):1317-23. [PubMed ID: 22500588]. https://doi.org/10.1111/j.1464-410X.2012.11133.x.

  • 5.

    Nokhandani AM, Otaghsara SMT, Abolfazli MK, Karimi M, Adel F, Babapour H, et al. A review of new method of cold plasma in cancer treatment. Scholars Acad J Biosci. 2015;3:222-30.

  • 6.

    Ermolaeva SA, Sysolyatina EV, Kolkova NI, Bortsov P, Tuhvatulin AI, Vasiliev MM, et al. Non-thermal argon plasma is bactericidal for the intracellular bacterial pathogen Chlamydia trachomatis. J Med Microbiol. 2012;61(Pt 6):793-9. [PubMed ID: 22361459]. https://doi.org/10.1099/jmm.0.038117-0.

  • 7.

    Stoffels E, Sakiyama Y, Graves DB. Cold Atmospheric Plasma: Charged Species and Their Interactions With Cells and Tissues. IEEE Transact Plasma Sci. 2008;36(4):1441-57. https://doi.org/10.1109/tps.2008.2001084.

  • 8.

    Rupf S, Lehmann A, Hannig M, Schafer B, Schubert A, Feldmann U, et al. Killing of adherent oral microbes by a non-thermal atmospheric plasma jet. J Med Microbiol. 2010;59(Pt 2):206-12. [PubMed ID: 19910483]. https://doi.org/10.1099/jmm.0.013714-0.

  • 9.

    Klampfl TG, Isbary G, Shimizu T, Li YF, Zimmermann JL, Stolz W, et al. Cold atmospheric air plasma sterilization against spores and other microorganisms of clinical interest. Appl Environ Microbiol. 2012;78(15):5077-82. [PubMed ID: 22582068]. https://doi.org/10.1128/AEM.00583-12.

  • 10.

    Isbary G, Morfill G, Schmidt HU, Georgi M, Ramrath K, Heinlin J, et al. A first prospective randomized controlled trial to decrease bacterial load using cold atmospheric argon plasma on chronic wounds in patients. Br J Dermatol. 2010:no. https://doi.org/10.1111/j.1365-2133.2010.09744.x.

  • 11.

    Arndt S, Unger P, Wacker E, Shimizu T, Heinlin J, Li YF, et al. Cold atmospheric plasma (CAP) changes gene expression of key molecules of the wound healing machinery and improves wound healing in vitro and in vivo. PLoS One. 2013;8(11). e79325. [PubMed ID: 24265766]. https://doi.org/10.1371/journal.pone.0079325.

  • 12.

    Bogle MA, Arndt KA, Dover JS. Evaluation of plasma skin regeneration technology in low-energy full-facial rejuvenation. Arch Dermatol. 2007;143(2):168-74. [PubMed ID: 17309997]. https://doi.org/10.1001/archderm.143.2.168.

  • 13.

    Kalghatgi SU, Fridman G, Cooper M, Nagaraj G, Peddinghaus M, Balasubramanian M, et al. Mechanism of Blood Coagulation by Nonthermal Atmospheric Pressure Dielectric Barrier Discharge Plasma. IEEE Transact Plasma Sci. 2007;35(5):1559-66. https://doi.org/10.1109/tps.2007.905953.

  • 14.

    Lee HW, Kim GJ, Kim JM, Park JK, Lee JK, Kim GC. Tooth bleaching with nonthermal atmospheric pressure plasma. J Endod. 2009;35(4):587-91. [PubMed ID: 19345811]. https://doi.org/10.1016/j.joen.2009.01.008.

  • 15.

    Cheng X, Murphy W, Recek N, Yan D, Cvelbar U, Vesel A, et al. Synergistic effect of gold nanoparticles and cold plasma on glioblastoma cancer therapy. Journal of Physics D: Applied Physics. 2014;47(33):335402. https://doi.org/10.1088/0022-3727/47/33/335402.

  • 16.

    Cheng X, Sherman J, Murphy W, Ratovitski E, Canady J, Keidar M. The effect of tuning cold plasma composition on glioblastoma cell viability. PLoS One. 2014;9(5). e98652. [PubMed ID: 24878760]. https://doi.org/10.1371/journal.pone.0098652.

  • 17.

    Yan D, Talbot A, Nourmohammadi N, Cheng X, Canady J, Sherman J, et al. Principles of using Cold Atmospheric Plasma Stimulated Media for Cancer Treatment. Sci Rep. 2015;5:18339. [PubMed ID: 26677750]. https://doi.org/10.1038/srep18339.

  • 18.

    Leclaire C, Lecoq E, Orial G, Clement F, Bousta F, editors. Fungal decontamination by cold plasma: an innovating process for wood treatment. Braga (Portugal): COST Action IE0601/ESWM-International Conference. 2008. p. 5-7.

  • 19.

    Kalghatgi S, Kelly CM, Cerchar E, Torabi B, Alekseev O, Fridman A, et al. Effects of non-thermal plasma on mammalian cells. PLoS One. 2011;6(1). e16270. [PubMed ID: 21283714]. https://doi.org/10.1371/journal.pone.0016270.

  • 20.

    Tsujimoto Y, Shimizu S. Role of the mitochondrial membrane permeability transition in cell death. Apoptosis. 2007;12(5):835-40. [PubMed ID: 17136322]. https://doi.org/10.1007/s10495-006-0525-7.

  • 21.

    Tuhvatulin AI, Sysolyatina EV, Scheblyakov DV, Logunov DY, Vasiliev MM, Yurova MA, et al. Non-thermal Plasma Causes p53-Dependent Apoptosis in Human Colon Carcinoma Cells. Acta Naturae. 2012;4(3):82-7. [PubMed ID: 23150806].

  • 22.

    Yan X, Zou F, Zhao S, Lu XP, He G, Xiong Z, et al. On the Mechanism of Plasma Inducing Cell Apoptosis. IEEE Transact Plasma Sci. 2010;38(9):2451-7. https://doi.org/10.1109/tps.2010.2056393.

  • 23.

    Volotskova O, Hawley TS, Stepp MA, Keidar M. Targeting the cancer cell cycle by cold atmospheric plasma. Sci Rep. 2012;2:636. [PubMed ID: 22957140]. https://doi.org/10.1038/srep00636.

  • 24.

    Vandamme M, Robert E, Lerondel S, Sarron V, Ries D, Dozias S, et al. ROS implication in a new antitumor strategy based on non-thermal plasma. Int J Cancer. 2012;130(9):2185-94. [PubMed ID: 21702038]. https://doi.org/10.1002/ijc.26252.

  • 25.

    Ahn HJ, Kim KI, Kim G, Moon E, Yang SS, Lee JS. Atmospheric-pressure plasma jet induces apoptosis involving mitochondria via generation of free radicals. PLoS One. 2011;6(11). e28154. [PubMed ID: 22140530]. https://doi.org/10.1371/journal.pone.0028154.

  • 26.

    Panngom K, Baik KY, Nam MK, Han JH, Rhim H, Choi EH. Preferential killing of human lung cancer cell lines with mitochondrial dysfunction by nonthermal dielectric barrier discharge plasma. Cell Death Dis. 2013;4. e642. [PubMed ID: 23703387]. https://doi.org/10.1038/cddis.2013.168.

  • 27.

    Yan X, Xiong Z, Zou F, Zhao S, Lu X, Yang G, et al. Plasma-Induced Death of HepG2 Cancer Cells: Intracellular Effects of Reactive Species. Plasma Proc Polymers. 2012;9(1):59-66. https://doi.org/10.1002/ppap.201100031.

  • 28.

    Hanschmann EM, Godoy JR, Berndt C, Hudemann C, Lillig CH. Thioredoxins, glutaredoxins, and peroxiredoxins--molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling. Antioxid Redox Signal. 2013;19(13):1539-605. [PubMed ID: 23397885]. https://doi.org/10.1089/ars.2012.4599.

  • 29.

    Hanschmann EM, Lonn ME, Schutte LD, Funke M, Godoy JR, Eitner S, et al. Both thioredoxin 2 and glutaredoxin 2 contribute to the reduction of the mitochondrial 2-Cys peroxiredoxin Prx3. J Biol Chem. 2010;285(52):40699-705. [PubMed ID: 20929858]. https://doi.org/10.1074/jbc.M110.185827.

  • 30.

    Koritzer J, Boxhammer V, Schafer A, Shimizu T, Klampfl TG, Li YF, et al. Restoration of sensitivity in chemo-resistant glioma cells by cold atmospheric plasma. PLoS One. 2013;8(5). e64498. [PubMed ID: 23704990]. https://doi.org/10.1371/journal.pone.0064498.

  • 31.

    Kim JY, Kim SO, Wei Y, Li J. A flexible cold microplasma jet using biocompatible dielectric tubes for cancer therapy. Appl Phys Lett. 2010;96(20):203701. https://doi.org/10.1063/1.3431392.

  • 32.

    Ma RN, Feng HQ, Liang YD, Zhang Q, Tian Y, Su B, et al. An atmospheric-pressure cold plasma leads to apoptosis in Saccharomyces cerevisiae by accumulating intracellular reactive oxygen species and calcium. J Phys D Appl Phys. 2013;46(28):285401. https://doi.org/10.1088/0022-3727/46/28/285401.

  • 33.

    Bekeschus S, Kolata J, Winterbourn C, Kramer A, Turner R, Weltmann KD, et al. Hydrogen peroxide: A central player in physical plasma-induced oxidative stress in human blood cells. Free Radic Res. 2014;48(5):542-9. [PubMed ID: 24528134]. https://doi.org/10.3109/10715762.2014.892937.

  • 34.

    Zucker SN, Zirnheld J, Bagati A, DiSanto TM, Des Soye B, Wawrzyniak JA, et al. Preferential induction of apoptotic cell death in melanoma cells as compared with normal keratinocytes using a non-thermal plasma torch. Cancer Biol Ther. 2012;13(13):1299-306. [PubMed ID: 22895073]. https://doi.org/10.4161/cbt.21787.

  • 35.

    Tanaka H, Mizuno M, Ishikawa K, Nakamura K, Kajiyama H, Kano H, et al. Plasma-Activated Medium Selectively Kills Glioblastoma Brain Tumor Cells by Down-Regulating a Survival Signaling Molecule, AKT Kinase. Plasma Med. 2011;1(3-4):265-77. https://doi.org/10.1615/PlasmaMed.2012006275.

  • 36.

    Yan D, Sherman JH, Cheng X, Ratovitski E, Canady J, Keidar M. Controlling plasma stimulated media in cancer treatment application. Appl Phys Lett. 2014;105(22):224101. https://doi.org/10.1063/1.4902875.

  • 37.

    Lee JH, Om JY, Kim YH, Kim KM, Choi EH, Kim KN. Selective Killing Effects of Cold Atmospheric Pressure Plasma with NO Induced Dysfunction of Epidermal Growth Factor Receptor in Oral Squamous Cell Carcinoma. PLoS One. 2016;11(2). e0150279. [PubMed ID: 26919318]. https://doi.org/10.1371/journal.pone.0150279.

  • 38.

    Wang M, Holmes B, Cheng X, Zhu W, Keidar M, Zhang LG. Cold atmospheric plasma for selectively ablating metastatic breast cancer cells. PLoS One. 2013;8(9). e73741. [PubMed ID: 24040051]. https://doi.org/10.1371/journal.pone.0073741.

  • 39.

    Guerrero-Preston R, Ogawa T, Uemura M, Shumulinsky G, Valle BL, Pirini F, et al. Cold atmospheric plasma treatment selectively targets head and neck squamous cell carcinoma cells. Int J Mol Med. 2014;34(4):941-6.

  • 40.

    Keidar M, Walk R, Shashurin A, Srinivasan P, Sandler A, Dasgupta S, et al. Cold plasma selectivity and the possibility of a paradigm shift in cancer therapy. Br J Cancer. 2011;105(9):1295-301. [PubMed ID: 21979421]. https://doi.org/10.1038/bjc.2011.386.

  • 41.

    Kim JY, Ballato J, Foy P, Hawkins T, Wei Y, Li J, et al. Apoptosis of lung carcinoma cells induced by a flexible optical fiber-based cold microplasma. Biosens Bioelectron. 2011;28(1):333-8. [PubMed ID: 21820891]. https://doi.org/10.1016/j.bios.2011.07.039.

  • 42.

    Asano S, Urabe A, Okabe T, Sato N, Kondo Y. Demonstration of granulopoietic factor(s) in the plasma of nude mice transplanted with a human lung cancer and in the tumor tissue. Blood. 1977;49(5):845-52. [PubMed ID: 300638].

  • 43.

    Zhang X, Li M, Zhou R, Feng K, Yang S. Ablation of liver cancer cells in vitro by a plasma needle. Appl Phys Lett. 2008;93(2):21502. https://doi.org/10.1063/1.2959735.

  • 44.

    Walk RM, Snyder JA, Srinivasan P, Kirsch J, Diaz SO, Blanco FC, et al. Cold atmospheric plasma for the ablative treatment of neuroblastoma. J Pediatr Surg. 2013;48(1):67-73. [PubMed ID: 23331795]. https://doi.org/10.1016/j.jpedsurg.2012.10.020.

  • 45.

    Arndt S, Wacker E, Li YF, Shimizu T, Thomas HM, Morfill GE, et al. Cold atmospheric plasma, a new strategy to induce senescence in melanoma cells. Exp Dermatol. 2013;22(4):284-9. [PubMed ID: 23528215]. https://doi.org/10.1111/exd.12127.

  • 46.

    Georgescu N, Lungu CP, Lupu AR, Osiac M. Atomic Oxygen Maximization in High-Voltage Pulsed Cold Atmospheric Plasma Jets. IEEE Transact Plasma Sci. 2010;38(11):3156-62. https://doi.org/10.1109/tps.2010.2070811.

  • 47.

    Brulle L, Vandamme M, Ries D, Martel E, Robert E, Lerondel S, et al. Effects of a non thermal plasma treatment alone or in combination with gemcitabine in a MIA PaCa2-luc orthotopic pancreatic carcinoma model. PLoS One. 2012;7(12). e52653. [PubMed ID: 23300736]. https://doi.org/10.1371/journal.pone.0052653.

  • 48.

    Leduc M, Guay D, Leask RL, Coulombe S. Cell permeabilization using a non-thermal plasma. N J Phys. 2009;11(11):115021. https://doi.org/10.1088/1367-2630/11/11/115021.

  • 49.

    Keidar M. Plasma for cancer treatment. Plasma Sources Sci Technol. 2015;24(3):33001. https://doi.org/10.1088/0963-0252/24/3/033001.

  • 50.

    Volotskova O, Shashurin A, Stepp MA, Pal-Ghosh S, Keidar M. Plasma-Controlled Cell Migration: Localization of Cold Plasma− Cell Interaction Region. Plasma Med. 2011;1(1).