Biomolecules & Therapeutics 2023; 31(5): 496-514  https://doi.org/10.4062/biomolther.2023.027
Current Status and Future Trends of Cold Atmospheric Plasma as an Oncotherapy
Xiaofeng Dai1,2,*, Jiale Wu1, Lianghui Lu1 and Yuyu Chen1
1The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
2Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
*E-mail: xiaofeng.dai@jiangnan.edu.cn
Tel: +19906244386
Received: February 14, 2023; Revised: April 12, 2023; Accepted: April 25, 2023; Published online: September 1, 2023.
© The Korean Society of Applied Pharmacology. All rights reserved.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Cold atmospheric plasma (CAP), a redox modulation tool, is capable of inhibiting a wide spectrum of cancers and has thus been proposed as an emerging onco-therapy. However, with incremental successes consecutively reported on the anticancer efficacy of CAP, no consensus has been made on the types of tumours sensitive to CAP due to the different intrinsic characteristics of the cells and the heterogeneous design of CAP devices and their parameter configurations. These factors have substantially hindered the clinical use of CAP as an oncotherapy. It is thus imperative to clarify the tumour types responsive to CAP, the experimental models available for CAP-associated investigations, CAP administration strategies and the mechanisms by which CAP exerts its anticancer effects with the aim of identifying important yet less studied areas to accelerate the process of translating CAP into clinical use and fostering the field of plasma oncology.
Keywords: Cold atmospheric plasma, Cancer, Device, Research status, Trends
INTRODUCTION

As a rapidly developing and emerging discipline, physical plasma has been widely applied in medicine, electronics, industry, and military and daily life, and its applications in the medical sector have attracted much attention (Von Woedtke et al., 2019). Gas plasma was first proposed by the British physicist Crockes in 1879 as the ‘fourth state of matter’ (Hoffmann et al., 2013) and was named ‘plasma’ in 1929 by American scientists Langmuir and Tonks (Haertel et al., 2014). Cold atmospheric plasma (CAP), partially ionized gases produced at room temperature and atmospheric pressure, is composed of reactive oxygen and nitrogen species (RONS), such as hydroxyl radicals (OH•), singleton oxygen (O), superoxide (O2–), nitric oxide (NO•), hydrogen peroxide (H2O2), ozone (O3), nitrogen dioxide (NO2), nitrogen trioxide (NO3), dinitrogen tetroxide (N2O4), and nitrite in the form of anions or protons (OONO, ONOOH) (Davalli et al., 2018; Xiang et al., 2018; Dai et al., 2020a). In addition to these active substances, CAP also subjects cells to reactive macromolecules, electromagnetic fields, and ions (Tornin et al., 2021). One unique feature of CAP is its multimodality, i.e., its effect relies on a carefully designed cocktail of multiple reactive species that is not dependent on any single component and exhibits differential therapeutic outcomes depending on the dosing and ejection parameter configuration. In addition to biological alterations, CAP also causes chemical and physical alterations to cells by introducing mild radiation and thermal and electromagnetic effects.

The use of CAP in the medical sector date back to the mid-20th century (Laroussi, 2018) for wound healing, blood coagulation, ulcer prevention, decontamination, and surface purification (Hong et al., 2009; Gay-Mimbrera et al., 2016). The anticancer potential of CAP was first reported in 2007 when treating melanoma (Yan et al., 2017c; Zhou et al., 2020). Since then, there have been consecutive reports of success using CAP to selectively induce programmed death events such as apoptosis (Xiang et al., 2018), necrosis (Hoffmann et al., 2013), autophagy (Liu et al., 2022) and immunogenic cell death (ICD) (Van Loenhout et al., 2019); to halt tumour cell migration (Wang et al., 2021); to rewire malignant cell metabolism; and to arrest cancer stemness (Dai et al., 2022), the processes of which involve both genetic and epigenetic alterations (Dai et al., 2020a; Gangemi et al., 2022). The anticancer efficacy of CAP has been demonstrated in approximately 30 tumours, including breast cancer (Xiang et al., 2018; Dai et al., 2022), glioma (Yang et al., 2021), pancreatic cancer (Van Loenhout et al., 2019), bladder cancer (Wang et al., 2021), prostate cancer (Hua et al., 2021), and liver cancer (Liu et al., 2022). In addition to using various in vitro and in vivo cell or animal models, the first clinical trial of CAP as a cancer therapy was approved by the U.S. Food & Drug Administration (FDA) in July 2019 and completed in April 2021 (NCT04267575) (Mark, 2019; Zhou et al., 2020), wherein 17 out of 20 recruited stage IV malignant cancer patients remained alive by the end of the trial, demonstrating the safety and efficacy of CAP as a new technology for cancer treatment.

Given our incremental understanding of the roles and complexity of redox modulation in cancer and health (Dai et al., 2019; Zhou et al., 2020), increasing attention has been paid to plasma-associated precision medicine. However, without comprehensive elucidation of the underlying molecular mechanism, the successful clinical translation of CAP is limited. Thus, identifying the underlying research gaps by reviewing the current research status of this field will be of fundamental importance before plasma medicine can truly benefit human oncology and cancer patients.

In this paper, by reviewing tumour types and stages in which the efficacy of CAP has been explored, the varied experimental models used, and the cancer hallmarks with which CAP has been shown to be capable of interfering, we systematically classify current CAP-based oncological studies into carefully designed logical frameworks and identify possible research directions worthy of intensive investigation towards leveraging the unique strengths of CAP for effective cancer treatment.

CURRENT STATUS OF CAP AS AN ONCOTHERAPY

CAP as an oncotherapy classified by tumour type

Ever since the first report of the anticancer effect of CAP in 2007, its selectivity against cancer cells has been reported in approximately 30 types of tumours, with the most intensively investigated types being melanoma (Yan et al., 2021) and breast cancer (Xiang et al., 2018), which represent 16.5% and 14.5% of all studies, respectively (Table 1). The investigated tumour types are primarily distributed in eight systems: abdominal organs (22.5%), reproductive organs (22%), skin (20%), brain (15%), bone (9%), blood (6%), head (4.5%), and soft tissue (1%) (Fig. 1). Abdominal cancers represent a large proportion of solid tumours, and examination of their efficacy has attracted much attention. Cancers originating from the reproductive system are diverse, most of which are well characterized, offering hints to systematically decipher the molecular mechanism driving the anticancer properties of CAP. Many studies have explored the therapeutic potential of CAP in skin cancer, as CAP has long been used for skin care (Busco et al., 2020). Furthermore lesions on the skin can be easily reached even if the electromagnetic effect from CAP can only penetrate glass/polystyrene barriers as thick as 7 mm (Yan et al., 2021). Tumours residing in the craniocerebral system are difficult to treat and require new therapeutic approaches, to which CAP may be particularly well suited.

Figure 1. Cancer types and proportions in which cold atmospheric plasma (CAP) is being explored as an oncotherapy. As of 14 December 2022, the field of plasma oncology encompasses eight broad categories of cancer: abdominal organs (45 studies), reproductive organs (44 studies), skin (40 studies), brain (30 studies), bone (18 studies), blood (12 studies), head (9 studies), and soft tissue (2 studies). Breast cancer (‘reproductive organs’), melanoma (‘skin’), glioma (‘brain’), and osteosarcoma (‘bone’) were the most commonly investigated (Table 1).

Among the abdominal tumours that have been investigated (i.e., pancreatic cancer (Van Loenhout et al., 2019), lung cancer (Wang et al., 2022b), colorectal cancer (Schneider et al., 2018a), colon cancer (Huang et al., 2022), bladder cancer (Wang et al., 2021), hepatocellular carcinoma (Wang et al., 2022), cholangiocarcinoma (Vaquero et al., 2020), and oesophageal cancer (Estarabadi et al., 2021)), pancreatic cancer, one of the most aggressive solid tumours, has been the best studied (10 out of 45, Table 1). For tumours originating from the reproductive system (i.e., breast cancer (Xiang et al., 2018; Dai et al., 2022), prostate cancer (Hua et al., 2021), cervical cancer (Kim et al., 2009), ovarian cancer (Koensgen et al., 2017)), breast cancer are the most commonly studied (29 out of 44, Table 1). Among the different breast cancer subtypes, triple-negative breast cancers (TNBC, without surface expression of characteristic receptors, i.e., oestrogen receptor, progesterone receptor, human epidermal growth factor receptor 2) exhibit the greatest cancer stemness and lack effective therapeutic options (Dai et al., 2016). CAP has been shown to be capable of selectively triggering apoptosis (Xiang et al., 2018) and inhibit migration (Wang et al., 2021) in TNBCs, effects that were both attributed to the ability of CAP to arrest cancer stemness (Dai et al., 2022). Tumours thus far examined in the skin system primarily include melanoma (Kim et al., 2009; Yan et al., 2021), cutaneous squamous cell carcinoma (Wang et al., 2019) and basal cell carcinoma (Yang et al., 2020b), with melanoma being the most frequently reported (33 out of 40, Table 1). For example, by combining CAP with gold nanoparticles bound to anti-FAK antibodies, five times more melanoma cells were killed than by using CAP alone (Kim et al., 2009). Among cancers in the craniocerebral system with demonstrated responsive to CAP treatment (i.e., glioma (Van Loenhout et al., 2021), vestibular schwannoma (Yoon et al., 2018), neuroblastoma (Walk et al., 2013), and retinoblastoma (Silva-Teixeira et al., 2021)), glioma is best studied (24 out of 30, Table 1). CAP has been synergized with auranofin to trigger cell death and immunogenic responses in glioblastoma and has been combined with gold nanoparticles to treat glioblastoma, where an overall 30% increase in cell death was achieved compared with CAP alone (Cheng et al., 2014a).

CAP as oncotherapy classified by experimental models

Most investigations on the safety and efficacy of CAP as a selective approach against cancer cells rely on in vitro studies and/or small in vivo animal models, with relatively little effort devoted to other models such as organoids, clinical case studies or trials. Currently, no large animals, such as primates or pigs, have been used to assess the onco-therapeutic efficacy and safety of CAP.

The in vitro antitumour efficacy of CAP has been examined using petri dishes or porous plates for almost 30 types of cancer cells, including brain cancer, skin cancer, breast cancer, colorectal cancer, lung cancer, cervical cancer, leukaemia, liver cancer and head and neck cancer (Keidar et al., 2011; Kaushik et al., 2013; Wang et al., 2013; Zhao et al., 2013; Bavelloni et al., 2015; Xiang et al., 2018). In vitro studies have been extensively used to evaluate the selectivity of CAP against tumour cells and to explore its mechanism of action. For instance, CAP-treated mouse neuroblastoma cell cultures exhibit reduced metabolic activity and increased apoptosis (Walk et al., 2013). Lung cancer cells and mouse melanoma cells exhibit dose-dependent cytopenia in response to CAP (Keidar et al., 2011). Melanoma exhibited significantly increased cellular mortality compared with normal keratinocytes after being treated with CAP, suggesting the role of CAP-imposed RONS in inducing cellular lipid peroxidation and DNA damage that pushes cancer cells past the apoptotic threshold sooner than their healthy peers (Zucker et al., 2012). In studies using 17 cell lines covering 7 tumour types (lung cancer, liver cancer, colon cancer, adenocarcinoma, melanoma, oral carcinoma, and uterine sarcoma) and 2 types of normal cells (i.e., adipose-derived stem cells and lung fibroblasts), CAP selectively induced the apoptosis of p53-deficient cancer cells without harming their healthy peers, suggesting the role of CAP in accelerating the genome instability of malignant cells (Ma et al., 2014). A more in-depth investigation reported the role of AQP3 in mediating cellular entry of some CAP-imposed reactive species and its competition with FOXO1 in sharing the same E3 ubiquitin ligase for K48-ubiquitination, where FOXO1 was responsible for fostering the stemness of cancer cells (Dai et al., 2022).

Organoids, self-organized three-dimensional (3D) tissue cultures derived from stem cells, can be crafted to replicate the complexity of an organ. Although commercially available organoid models are limited, they have been used in several studies as 3D experimental models to examine the anticancer efficacy of CAP. For instance, CAP demonstrated selective toxicity against bladder cancer cells in patient-derived preclinical organoid models (Gelbrich et al., 2023) and reduced the spheroid area and halted epithelial-mesenchymal transition (EMT) in patient-derived TNBC and bladder cancer organoids (Wang et al., 2021). Additionally, cytotoxicity, spheroid shrinkage and cell migration were monitored using 3D glioblastoma spheroid tumours after CAP treatment, where the impact of short-lived species was emphasized (Privat-Maldonado et al., 2018).

In vivo animal studies have largely relied on mouse models, including in situ models, subcutaneous xenograft models, peritoneal models and intracranial models (Chen et al., 2017). In vivo small animal assays have primarily been used to confirm the onco-therapeutic efficacy of CAP and the perturbed signalling networks identified using in vitro studies. After hypothesizing that CAP exhibits selective toxicity towards cancer cells, researchers used in vivo murine models of subcutaneous bladder tumours and melanoma to demonstrate that CAP can suppress tumour growth, enhance tumour shrinkage, and improve mouse survival (Keidar et al., 2011). Using a mouse neuroblastoma model, CAP was shown to be capable of ablating tumours in vivo, where the median survival duration was prolonged from 15 to 28 days (p<0.001) (Walk et al., 2013). In addition to arresting tumour growth, CAP reduced the migration of TNBCs in vivo, as demonstrated by enhanced expression of E-cadherin, a suppressive marker of metastasis (Zhou et al., 2020). Several studies have also explored the synergistic effects of CAP with therapeutics such as traditional chemotherapies and targeted agents using animal models. For instance, CAP exhibited enhanced selective cytotoxicity against melanoma when combined with dacarbazine (Alimohammadi et al., 2020) or doxorubicin (Pefani-Antimisiari et al., 2021) in vivo. A combinatorial strategy taking advantage of CAP and paclitaxel resulted in enhanced death and a higher frequency of DNA fragmentation than using paclitaxel alone in breast cancer (Mihai et al., 2022).

Several clinical case studies and clinical trials have been launched to prove the feasibility of CAP in cancer treatment. CAP has been used as a catheter-guided scalpel for surgery, with a significant postoperative curative effect reported for head and neck cancers. Specifically, 12 patients with head and neck cancers were recruited, with superficial partial tumour remission, healing of infected ulcerations, and decreased pain medication requirements after CAP treatment (Metelmann et al., 2015). Later, 6 patients with locally advanced squamous cell carcinoma of the oropharynx suffering from open infected ulcerations were recruited, and partial tumour remission in response to CAP was reported for 2 patients; the efficacy of CAP was attributed to CAP-induced ICD (Metelmann et al., 2018). Cold plasma scalpel CHCPS functioned as an adjunct to intraoperative radiotherapy in removing intraperitoneal sarcoma (Isbary et al., 2010). The Canady scalpel, a tool of CAP established for cancer treatment, was coupled with surgery to treat a 33-year-old with rare recurrent peritoneal sarcoma in 2019 (2020). In a clinical trial examining the efficacy of CAP against actinic keratotic lesions (NCT02759900), 19 out of 24 patients exhibited remission of their lesions, thereby decreasing the development of squamous cell carcinoma (the transformation rate of intraepidermal keratinocytic dysplasia to invasive squamous cell carcinoma is approximately 20% within 10-25 years) (Friedman et al., 2017; Wirtz et al., 2018). The first clinical trial using CAP as an oncotherapy was initiated in 2019 and completed in 2021 and involved 20 stage IV late-stage cancer patients (NCT04267575) (Mark, 2019; Zhou et al., 2020).

CAP as an oncotherapy classified by treatment approaches

CAP can be delivered to cells through both direct and indirect approaches. Direct treatment refers to exposing cells or tumours directly to plasmas ejected from various plasma sources. Indirect strategies typically treat objects with plasma-activated medium (PAM), such as plasma-activated normal saline (PSW) or plasma-activated PBS solution (PAPBS) (Chen et al., 2017) (Fig. 2).

Figure 2. Diagram summarizing differences between direct CAP injection and indirect PAM treatment.

For direct treatment, there are three main ways to source CAP (Bernhardt et al., 2019): dielectric barrier discharge (DBD), plasma jet, and plasma torch (Dai et al., 2018). The most striking difference between DBD and plasma jet or plasma torch lies in that the tissues or cells being treated act as electrodes in DBD but not in the other two approaches (Yan et al., 2017c). In addition, plasmas ejected from DBD and jet/torch differ in shape and range. Although plasma ejected by jet/torch is slender, has no fixed boundary, has a small range of action (Xu et al., 2021) and can be used for medical applications, DBD is advantageous in handling 3D objects and covers large surface areas (Corbella et al., 2021). It is noteworthy that the plasma jet exhibited greater efficacy in triggering the death of acute myeloid leukaemia cells than did DBD under the same conditions (Xu et al., 2021).

Among these strategies, DBD was the earliest approach and was proposed by Siemens and his colleagues in 1857 (Hoffmann et al., 2013). As a commercial example of DBD, the PlasmaDerm® series (Fig. 3A) has been widely used to promote chronic wound healing and treat chronic inflammatory diseases (Bernhardt et al., 2019; Wandke et al., 2022). Koinuma et al. (1992) developed the first cold plasma jet in 1992. Plasma jets have been widely applied for medical purposes (Stoffels et al., 2002; Laroussi and Lu, 2005; Laroussi et al., 2006; Breathnach et al., 2018), with kINPen® being the most widely used (Isbary et al., 2013b) (Fig. 3B). Microwave-driven plasma torches have also been launched for disease treatment, such as the use of microPlaSterβ® (Fig. 3C) in wound healing (Arndt et al., 2013). Several miniature plasma jets, such as invivoPen and µCAP, have been established and used in cancer treatment (Chen et al., 2017; Zhou et al., 2020) (Fig. 3D, 3E). In addition, the activity of CAP can be delivered in liquid form, i.e., PAM, which can be easily stored, transported, and administered, largely expanding its application scenarios (Sersenova et al., 2021).

Figure 3. Representative cold atmospheric plasma (CAP) sources. Images and schematic presentations of (A) single electrode DBD, PlasmaDerm® VU-2010 (CINOGY System GmbH plasma technology, Duderstadt, Germany); (B) plasma jet, kINPen® (INP Greifswald/neoplas GmbH, Greifswald, Germany); (C) plasma torch, microPlaSterβ® (Adtec Plasma Technology Co. Ltd., London, UK); and plasma sources for in vivo treatment, i.e., (D) invivoPen and (E) µCAP plasma generator. (A) was reproduced with permission from Bernhardt et al. (2019) under a Creative Commons CC-BY licence; (B) was reproduced with permission from Breathnach et al. (2018) under a Creative Commons CC-BY licence; (C) was reproduced with permission from Arndt et al. (2013a) under a Creative Commons CC-BY licence; (D) was reproduced with permission from Zhou et al. (2020) under a Creative Commons CC-BY licence; and (E) was reproduced with permission from Chen et al. (2017) under a Creative Commons CC-BY licence.

Direct ejection: In principle, direct CAP ejection exhibits anticancer efficacy comparable to that of indirect PAM treatment despite the observation that direct CAP treatment slightly raised the temperature of cells by approximately 1-2°C (Keidar et al., 2011).

Most CAP sources require direct cell or tissue exposure to deliver their active species and have limited penetration. Thus, direct CAP treatment has been largely used in in vitro assays, for skin cancers in vivo, and combined with surgery in clinical studies. By applying a convenient and portable ambient air-fed device, namely, aCAP, for local postoperative cancer treatment targeting residual tumour cells in the surgical cavity, CAP induced ICD and antitumour immunity by releasing tumour-associated antigens (TAAs) in situ. Immature dendritic cells (DCs) phagocytose TAAs and process them into peptides during the migration of TAAs to the tumour-draining lymph nodes. Here, mature DCs present antigenic peptides to T cells, producing cytotoxic T cells that inhibit tumour recurrence and suppress tumour growth. Treating residual tumour cells in the operative cavity with aCAP after surgical resection of 4T1 breast tumours significantly increased the survival of mice compared with the control group (Chen et al., 2021). Direct CAP treatment of melanoma reduced tumour burden and prolonged mouse survival by enhancing the cancer-immunity cycle (Lin et al., 2022). The Canady scalpel has been successful in treating patients with pancreatic cancer or rare recurrent peritoneal sarcoma (2020) and had been used in conjunction with surgery in a clinical trial involving 20 late-stage malignant cancer patients (NCT04267575) (Mark, 2019; Zhou et al., 2020).

Despite the demonstrated feasibility of CAP as an onco-therapy, direct CAP exposure can only cause cell death in the upper 3 to 5 cell layers (Chen et al., 2017). A novel plasma jet capable of ejecting CAP within tissues, namely, invivoPen, has been established and exhibits efficacy similar to PAM in treating TNBC in mice, suggesting the possibility of achieving minimally invasive cancer surgery (Zhou et al., 2020). Traditional CAP devices are too large for treating intracranial cancers. A micro-CAP device (μCAP) was invented to deliver plasma through an intracranial endoscopic tube to target glioblastoma in mice. The approach exhibited improved therapeutic outcomes compared with indirect approaches with a reduced gas flow rate and plasma jet size (Chen et al., 2017).

Indirect treatment: Unlike direct CAP treatment, which requires direct contact with cells/tissues and is restricted by limited depth penetration, the reactive species of CAP can be stored in the form of medium (i.e., PAM) and preserved for up to a week. Thus, PAM has been widely applied in CAP-based cancer investigations, especially in treating tumours residing in deep tissues, intraperitoneal tumours and metastatic tumours (Chen et al., 2017; Kaushik et al., 2018; Keidar et al., 2018). However, the efficacy of PAM may be reduced due to the nutrients it contains. Foetal bovine serum (FBS) in whole cell medium and pyruvate in Dulbecco’s modified Eagle’s medium (DMEM) can reduce the anticancer properties of PAM (Yan et al., 2016b). In addition, reactive species in PAM degrade over time due to the reaction of CAP substances with cell medium components (primarily cysteine and methionine), which can be significantly mitigated by the addition of 3-nitro-L-tyrosine or sequestration at –80°C (Yan et al., 2016a). The incubation time and dilution of PAM during treatment also affect the efficacy of PAM (Chen et al., 2017; Kaushik et al., 2018; Zhou et al., 2020). Many parameters may affect the efficacy of PAM, complicating its dosing. For instance, the use of larger wells in multiwell plates, shorter distances between the plasma source and the medium, and smaller media volumes have been reported to promote the anticancer properties of PAM, as the anticancer effect is determined by the amount of PAM acting on individual cells rather than the therapeutic dose of PAM as a whole (Yan et al., 2015; Jo et al., 2022a).

PAPBS and PAW are plasma-treated buffer solutions and differ from PAM in lacking the essential nutrients cells required to survive. However, PAPBS and PAW only affect cells for short periods of time (i.e., several hours), which has substantially restricted their application. For example, unlike PAM, which has demonstrated significant anticancer effects on a variety of cancer cells, PAW has no obvious cytotoxic effect on glioblastoma (Chen et al., 2017). This finding underlines the importance of establishing novel technologies capable of restoring the activity of PAM. Among various RONS, hydrogen peroxide has been proposed to play the critical role in the selectivity of CAP against cancer cells (Dai et al., 2022). Encouragingly, plasma-activated hydrogel (PAH), prepared by directly dissolving acrylamide, polymerizing acrylamide in PAW, and supplementing the mixture with crosslinking agents, initiators, and polymerization inhibitors, has been shown to prolong the lifespan of hydroxyl radicals for up to 140 days (Liu et al., 2019). However, the anticancer effect of CAP is complemented by other reactive species (Bauer et al., 2019; Dai et al., 2022), including singlet oxygen, which has been reported to play a leading role in selectively inducing cancer cell apoptosis (Sersenova et al., 2021). Effective strategies for preserving the activities of these species remain unknown.

CAP as an oncotherapy classified by molecular mechanisms

Both direct and indirect treatments can selectively induce the death of cancer cells by elevating cellular RONS levels. One prevailing explanation suggests that cancer cells exhibit higher cellular RONS levels and a more fragile antioxidant system than normal cells, rendering cancer cells more sensitive to redox stress and thus closer to the death threshold (Gorrini et al., 2013; Dai et al., 2022). Another theory attributes the selectivity of CAP against cancer cells to the lower levels of cholesterols and/or more aquaporins in cancer cell membranes, which are associated with increased RONS intracellular flow (Van der Paal et al., 2017).

With these conceptual explanations of the anticancer efficacy of CAP, it is necessary to identify the specific pathways and cancer hallmarks they target to promote the establishment of a CAP-based precision oncological system. CAP is known to interfere with all of the 10 well-recognized cancer hallmarks (Senga and Grose, 2021). These hallmarks can be groups into five traits: selective triggering of programmed death, halting tumour angiogenesis and metastasis, rewiring tumour metabolism, boosting immunity and suppressing tumour-promotive inflammation, and accelerating cancer cell genome instability (Fig. 4).

Figure 4. Effects of cold atmospheric plasma (CAP) on cancer hallmarks. The 10 cancer hallmarks (Senga and Grose, 2021) can be summarized into five traits, i.e., selective triggering of programmed death events in cancer cells, halting of tumour angiogenesis and metastasis, rewiring of tumour metabolism, boosting of immunity and suppression of tumour-promoting inflammation, and accelerated cancer cell genome instability. In response to reactive oxygen and nitrogen species (RONS) induced by CAP, the genome stability of cancer cells is compromised, which accelerates the mutation rate. As the genome integrity of cancer cells is associated with cancer stemness and drug resistance, targeting the integrity of transformed cells often leads to the killing of cancer stem cells and enhanced sensitivity of cancer cells to oncoagents. Cellular RONS levels continue to increase with cell chaos as a result of accelerated genome mutation that, once exceeding the death threshold as sensed by p53 (e.g.), may trigger various types of programmed cell death events. On the other hand, mitochondria, in response to enhanced cellular RONS, may trigger death programs by releasing, e.g., cytochrome C, which activates caspases. CAP can reshape cancer cell metabolism, including that of carbohydrates, nucleic acids, lipids, and amino acids, by redirecting pyruvate from increasing lactate production to enhancing efficient energy production, enabling β-oxidation and disturbing amino acid metabolism. In addition, CAP can promote tumour antigen release, stimulate immunogenic cell death (ICD), enhance tumour antigen presentation by antigen presentation cells, and enhance the sensitivity of tumour cells to immune therapies. Finally, CAP can arrest cancer angiogenesis and metastasis by halting the epithelial-mesenchymal transition (EMT).

Programmed cell death: CAP is capable of selectively inducing a wide panel of programmed cell death events in cancer cells, including apoptosis, cell cycle arrest, autophagic cell death, ferroptosis, pyroptosis, necroptosis, and ICD. For example, CAP induced apoptosis in human colon and lung cancer cells (Wang et al., 2022b), selectively induced G0/G1 cell cycle arrest in androgen receptor (AR)-independent prostate cancer cells (Hua et al., 2021), induced the expression of autophagy-related genes in melanoma cells (Alimohammadi et al., 2020), triggered human lung cancer cell ferroptosis by enhancing intracellular and lipid ROS levels (Jo et al., 2022a), induced necroptosis in vestibular schwannoma cells (Yoon et al., 2018), initiated ICD in colorectal cancers in vivo (Lin et al., 2018), and led to pyroptosis in a dose-dependent manner in lung, gastric, and hepatoma carcinoma cells (Yang et al., 2020a).

With an increased understanding of cell death programs, novel cell death events continue to be identified. Cuproptosis, a copper-triggered modality of mitochondrial cell death (Wang et al., 2022c), has recently attracted much attention. Given the significant overlap between cuproptosis and ferroptosis with respect to gene signatures and molecular mechanisms, it is highly possible that CAP also triggers cuproptosis, which warrants intensive investigation.

Tumour angiogenesis and metastasis: CAP plays an inhibitory role in tumour angiogenesis by selectively inhibiting VEGFA expression and altering the tumour microenvironment (TME) in vivo. For instance, CAP significantly reduced intratumoural vascular density in hepatocellular carcinoma and weakened microcirculation by causing vascular occlusion in the tumour-associated vascular system established using a hepatocellular carcinoma model (Kugler et al., 2022). It is worth noting that while inhibiting tumour angiogenesis, CAP can improve wound angiogenesis by activating angiogenesis-related healthy cells such as desquamated cells, fibroblasts and endothelial cells (Dai et al., 2020a).

CAP halts cancer cell migration and invasion by interfering with the epithelial mesenchymal transition (EMT). Specifically, although CAP has been shown to be capable of inducing epithelial cell apoptosis (Dezest et al., 2017), it preferentially inhibited the growth of mesenchymally shifted carcinoma cells without harming their epithelial peers in bladder and breast cancer organoid models (Wang et al., 2021). CAP, by synergizing with temozolomide, reduced glioblastoma cell migration by increasing the cell surface expression of αvβ3 and αvβ5 integrins (Gjika et al., 2020). Additionally, CAP inhibited the migration of myeloma cells by decreasing the secretion of MMP-2 and MMP-9, both of which are key metalloproteinases for degrading the extracellular matrix (Xu et al., 2016).

Tumour metabolism: CAP can rewire the metabolism of cancer cells by primarily perturbing carbohydrate metabolism, lipid metabolism, amino acid metabolism and energy metabolism.

Cancer cells are characterized by aerobic glycolysis, wherein glucose is converted into pyruvate by glucose transporters and glycolytic enzymes to produce sufficient biomass and adenosine triphosphate (ATP) for sustained cell proliferation. During this process, excess lactate is exported to the TME, which fosters an acidic environment favouring accelerated tumour growth. CAP can neutralize this acidic environment by inhibiting several major enzymes of glycolysis, including hexokinase (HK), phosphofructokinase-1 (PFK-1), pyruvate kinase (PK), and lactate dehydrogenase (LDH), to reduce the production of lactate (Guo et al., 2021). Once glucose enters cells, it is first catalysed by HK to generate glucose 6-phosphate, followed by catalysis by PFK-1, PK, LDH and other enzymes to produce energy. CAP blocks the glycolytic pathway by inhibiting hexokinase phosphorylation in blood cancer cells, which subsequently decreased cellular ATP levels and inhibited cancer cell proliferation (Kaushik et al., 2015). CAP downregulated glucose transporters (GLUTs) by suppressing PI3K/AKT signalling in glioblastoma cells (Makinoshima et al., 2014) and suppressed intracellular expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a classical glycolytic enzyme (Lackmann et al., 2013).

Another strategy adopted by cancer cells to enhance glycolysis and uncontrolled proliferation is the suppression of pentose phosphate signalling, wherein glucose is converted to nucleotides instead of lactate and energy. CAP can modify the metabolic profiles of U251SP glioblastoma cells by inhibiting the glycolytic pathway and enhancing pentose phosphate signalling (Kurake et al., 2019).

Lipids are key components of cells and the plasma membrane, which form an impermeable barrier and plays an important role in numerous cellular processes, such as cell growth, proliferation, differentiation, and signalling (Pan et al., 2021). CAP inhibited β-alanine metabolism in myeloma cells, leading to inhibition of fatty acids (FAs) synthesis by coenzyme A (CoA), as well as disrupted energy and material metabolism in cancer cells (Xu et al., 2018b).

Glutamate is an essential nutrient that sustains uncontrolled tumour cell growth and is produced through glutamine metabolism. CAP blocked the nutritional source of leukaemic cells by inhibiting the activity of glutaminase, thereby decreasing glutamate production (Xu et al., 2019b, 2021).

Tumour-promoting inflammation and immunity: DCs and macrophages in the innate immune system play essential roles in the balance between inflammation and anti-inflammation. The primarily role of DCs is to present TAAs to T cells to induce the production of tumour antigen-specific cytotoxic T cells (CTLs), which leads to activation of antitumour immunogenic programs. CAP synergizes with auranofin in activating DCs and initiating the antitumour immune response in glioblastoma cells (Van Loenhout et al., 2021). Additionally, CAP stimulated the differentiation of monocytes into macrophages (Kaushik et al., 2019) and primed macrophages towards the M1 state (exhibiting an antitumour phenotype) to promote an antitumour microenvironment by increasing the expression of cytokines associated with the M1 phenotype, including TNFα, TNFsf1 and Il1β. Cancer models used to explore these phenomena include human cholangiocarcinoma and glioma (Kaushik et al., 2019; Vaquero et al., 2020; Duchesne et al., 2021).

In adaptive immunity, CAP acted as a unique RONS delivery system to stimulate the immune system and promote an anticancer immune response (Bekeschus et al., 2017; Lin et al., 2019; Duchesne et al., 2021; Gangemi et al., 2022; Min et al., 2022). CAP reversed the downregulated immune response in glioblastoma multiforme (Almeida et al., 2019) and promoted the tumour recruitment of immune cells by releasing immune stimulation signals in the TME of pancreatic cancer cells (Lin et al., 2018; Almeida et al., 2019; Van Loenhout et al., 2019). CAP synergized with photodynamic therapy (PDT) in inducing greater apoptotic cell death in human papillomavirus (HPV)-positive cervical cancers than using PDT alone (Kim et al., 2009).

Genome stability: CAP sabotages the genome stability of cancer cells by causing DNA damage and interfering with the DNA repair system. Accelerated genome instability ultimately triggers death program due to rapid accumulation of damage that is intolerable by cells. CAP can induce DNA damage events in cancer cancers, especially double-strand breaks (DSBs), during the early stage of CAP treatment, which is marked by specific phosphorylation of threonine 139 on the H2A histone family member X (γ-H2AX). Recruitment of ataxia telangiectasia mutation (ATM) at the DSB threonine phosphorylation sites leads to subsequent p53 phosphorylation, DNA damage response complex formation, p21 activation and cell cycle arrest (Joerger and Fersht, 2008). In addition to cell cycle arrest, p53 phosphorylation activates proapoptotic factors such as Bax, Puma and Noxa, resulting in the release of cytochrome C into the cytoplasm and its binding with apoptotic protease factor-1 (Apaf-1). The subsequent formation of apoptotic bodies promotes cleavage-mediated activation of caspase-9 and caspase-3/7, and, ultimately, the initiation of apoptosis (Yan et al., 2017c; Chen et al., 2022). In addition to directly inducing DNA damage, CAP can impose endoplasmic reticulum (ER) redox stress, which indirectly endangers genome stability. During ER stress, Bax and Bak in the ER allow Ca2+ to be released from the ER into the cytoplasm, leading to m-Calpain and subsequent caspase-12 activation. The downstream sequential activation of caspase family members promotes the release of cytochrome C into the cytoplasm, apoptotic body formation, inhibition of anti-apoptotic factors and activation of pro-apoptotic proteins such as Bim, Bax, and Bak (Dai et al., 2021).

Given that CAP can promote genome instability, it is possible that CAP can arrest cancer stemness by altering the cellular antioxidant system. Although cancer stem cells (CSCs) have relatively lower redox levels relative to bulk tumour cells and noncancer cells, CSCs exhibit greater sensitivity to CAP treatment due to altered antioxidant homeostasis as a result of impeded genome stability (Dai et al., 2022). This theory has been examined in TNBC cells, which harbour a greater proportion of CSCs than other breast cancer subtypes. In TNBC cells, CAP treatment resulted in significant reductions in the levels of FOXO1 (antioxidant protein marker), ALDH1 (a marker of breast cancer stemness) and IL6 (a protein associated with cancer stemness) (Zhou et al., 2020; Sersenova et al., 2021; Dai et al., 2022).

Drug resistance is a common issue in oncology and is associated with the persistence of residual CSCs after surgery or drug therapy. Several experiments have demonstrated the ability of CAP to restore the drug sensitivity of cancer cells. For example, CAP rewired the resistance of 73% of breast cancer cells to paclitaxel by altering the expression of 49 genes, including Bcl2l13, Hoxa9, Dagla, and Ceacam1 (Park et al., 2019). CAP treatment also sensitized breast cancer cells to tamoxifen by altering the mRNA levels of Ma1 and Hoxc6 (Lee et al., 2017).

FUTURE TRENDS IN CAP AS AN ONCOTHERAPY

With our increasing understanding of the unique effects of CAP on cancer cells, CAP has emerged as an effective tool for preventing cancer metastasis and drug resistance by targeting cancer stemness (Dai et al., 2022). Since 2007, when the tumour selectivity of CAP was established, its effectiveness in selectively targeting cancer cells without harming their healthy peers has been demonstrated in diverse types of cancers (Xiang et al., 2018; Van Loenhout et al., 2019; Hua et al., 2021; Wang et al., 2021; Yang et al., 2021; Dai et al., 2022; Liu et al., 2022). These studies collectively suggest that CAP possess significant potential for clinical translation as a precision oncological tool, alone or by synergizing with other treatment modalities. For instance, CAP significantly inhibited the growth of head and neck cancers without obvious side effects in 2015 (Schuster et al., 2016), prolonged the lifespan of a 75-year-old patient with late-stage pancreatic cancer for 2 additional years in 2016, and altered the course of a 33-year-old patient with rare relapsed incurable peritoneal sarcoma in 2019 (2020). Despite these encouraging clinical efforts (Dai et al., 2018), several issues remain and require more focused attention.

Although a large panel of cancer types are responsive to CAP treatment, no systematic investigation have been conducted to identify features sensitizing cells to CAP treatment, nor have intensive efforts been made to establish a dosing system capable of arresting cancer cells based on their molecular characteristics. The lack of such a system may be due to different parameter configurations and the types of gases used to generating plasma in different studies, which results in differential combinations of reactive species and dosing intensities and renders these results difficult to compare. This system is of paramount importance before CAP can be accurately translated into clinical practice, as the outcome of CAP is dose-dependent (Dai et al., 2020b). A bridge between various CAP-initiated cell death programs and intrinsic cellular characteristics is required to achieve desirable therapeutic outcomes.

Current studies on CAP largely rely on in vitro assays. The establishment of organoids makes it possible to investigate the 3D behaviour of cells in response to CAP using an organ-mimicking system without resorting to in vivo animal experiments. In addition, organoid models make it possible to screen agents capable of improving the therapeutic efficacy of CAP, largely enhancing the ability to identify molecules that are likely to synergize with CAP. However, commercially available organoid tumour models remain limited, and relatively few studies have relied on organoids for CAP-associated investigations, both of which deserve intensified focus.

Direct plasma treatment, although superior to indirect approaches in cancer treatment, has difficulty reaching deep lesions. Although devices such as μCAP and invivoPen have been developed, investigations in this area are far from sufficient to resolve all issues. Indirect plasma treatment, although adding a great degree of flexibility to gas plasma administration, faces the obstacle of preserving the activity of PAM. I The finding that hydroxyl radicals, one of the leading reactive species in promoting cancer cell death (Dai et al., 2022), can be preserved in the form of PAHs for as long as 140 days is highly encouraging (Liu et al., 2019). However, the multimodal characteristic of CAP does not rely on any single component but requires the aid of other species, making the preservation of other reactive species equally important. This may represent a pivotal issue hampering the use of CAP in the medical sector.

Most studies on the anticancer mechanism of CAP focus on various types of death programs, migration and metastasis. With increasing evidence concerning the benefits of immunotherapy, greater attention has shifted to CAP-associated cancer immunity. However, relatively little has been devoted to deciphering the role of CAP in suppressing tumour angiogenesis, reshaping cancer metabolism, and accelerating cancer genome instability. The elucidation of these effects may deepen our understanding of the functionality of redox modulation in cancer initiation and progression. Additionally, CAP is known to trigger multiple death programs in cancer, the precise form of which varies with the components and dosing of CAP. As novel forms of programmed cell death and molecular mechanisms continue to emerge, it is interesting to examine whether CAP can potentiate these programs or possibly promote as of yet undescribed death events or pathways that are unique to CAP or redox modulation.

ACKNOWLEDGMENTS
This work was supported by the National Natural Science Foundation of China (Grant No. 81972789) and Fundamental Research Funds for the Central Universities (Grant No. JUSRP22011). The funding bodies played no role in the design of the study; collection, analysis, and interpretation of data; or in manuscript preparation.
CONFLICT OF INTEREST

The authors declare no competing interest.

References
  1. Adhikari, M., Adhikari, B., Ghimire, B., Baboota, S. and Choi, E. H. (2020) Cold atmospheric plasma and silymarin nanoemulsion activate autophagy in human melanoma cells. Int. J. Mol. Sci. 21, 1939.
    Pubmed KoreaMed CrossRef
  2. Adhikari, M., Kaushik, N., Ghimire, B., Adhikari, B., Baboota, S., Al-Khedhairy, A. A., Wahab, R., Lee, S.-J., Kaushik, N. K. and Choi, E. H. (2019) Cold atmospheric plasma and silymarin nanoemulsion synergistically inhibits human melanoma tumorigenesis via targeting HGF/c-MET downstream pathway. Cell Commun. Signal. 17, 52.
    Pubmed KoreaMed CrossRef
  3. Afrasiabi, M., Tahmasebi, G., Eslami, E., Seydi, E. and Pourahmad, J. (2022) Cold atmospheric plasma versus cisplatin against oral squamous cell carcinoma: a mitochondrial targeting study. Iran. J. Pharm. Res. 21, e124106.
    Pubmed KoreaMed CrossRef
  4. Aggelopoulos, C. A., Christodoulou, A.-M., Tachliabouri, M., Meropoulis, S., Christopoulou, M.-E., Karalis, T. T., Chatzopoulos, A. and Skandalis, S. S. (2022) Cold atmospheric plasma attenuates breast cancer cell growth through regulation of cell microenvironment effectors. Front. Oncol. 11, 826865.
    Pubmed KoreaMed CrossRef
  5. Akter, M., Jangra, A., Choi, S. A., Choi, E. H. and Han, I. (2020) Non-thermal atmospheric pressure bio-compatible plasma stimulates apoptosis via p38/MAPK mechanism in U87 malignant glioblastoma. Cancers (Basel) 12, 245.
    Pubmed KoreaMed CrossRef
  6. Alimohammadi, M., Golpur, M., Sohbatzadeh, F., Hadavi, S., Bekeschus, S., Niaki, H. A., Valadan, R. and Rafiei, A. (2020) Cold atmospheric plasma is a potent tool to improve chemotherapy in melanoma in vitro and in vivo. Biomolecules 10, 1011.
    Pubmed KoreaMed CrossRef
  7. Almeida-Ferreira, C., Silva-Teixeira, R., Gonçalves, A. C., Marto, C. M., Sarmento-Ribeiro, A. B., Caramelo, F., Botelho, M. F. and Laranjo, M. (2022) Cold atmospheric plasma apoptotic and oxidative effects on MCF7 and HCC1806 human breast cancer cells. Int. J. Mol. Sci. 23, 1698.
    Pubmed KoreaMed CrossRef
  8. Almeida, N. D., Klein, A. L., Hogan, E. A., Terhaar, S. J., Kedda, J., Uppal, P., Sack, K., Keidar, M. and Sherman, J. H. (2019) Cold atmospheric plasma as an adjunct to immunotherapy for glioblastoma multiforme. World Neurosurg. 130, 369-376.
    Pubmed CrossRef
  9. Ando, T., Suzuki-Karasaki, M., Suzuki-Karasaki, M., Ichikawa, J., Ochiai, T., Yoshida, Y., Haro, H. and Suzuki-Karasaki, Y. (2021) Combined anticancer effect of plasma-activated infusion and salinomycin by targeting autophagy and mitochondrial morphology. Front. Oncol. 11, 593127.
    Pubmed KoreaMed CrossRef
  10. Ara, E. S., Noghreiyan, A. V. and Sazgarnia, A. (2021) Evaluation of photodynamic effect of Indocyanine green (ICG) on the colon and glioblastoma cancer cell lines pretreated by cold atmospheric plasma. Photodiagnosis Photodyn. Ther. 35, 102408.
    Pubmed CrossRef
  11. Arndt, S., Fadil, F., Dettmer, K., Unger, P., Boskovic, M., Samol, C., Bosserhoff, A.-K., Zimmermann, J. L., Gruber, M., Gronwald, W. and Karrer, S. (2021) Cold atmospheric plasma changes the amino acid composition of solutions and influences the anti-tumor effect on melanoma cells. Int. J. Mol. Sci. 22, 7886.
    Pubmed KoreaMed CrossRef
  12. Arndt, S., Unger, P., Wacker, E., Shimizu, T., Heinlin, J., Li, Y. F., Thomas, H. M., Morfill, G. E., Zimmermann, J. L., Bosserhoff, A. K. and Karrer, S. (2013a) 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 8, e79325.
    Pubmed KoreaMed CrossRef
  13. Arndt, S., Wacker, E., Li, Y.-F., Shimizu, T., Thomas, H. M., Morfill, G. E., Karrer, S., Zimmermann, J. L. and Bosserhoff, A.-K. (2013b) Cold atmospheric plasma, a new strategy to induce senescence in melanoma cells. Exp. Dermatol. 22, 284-289.
    Pubmed CrossRef
  14. Azzariti, A., Iacobazzi, R. M., Di Fonte, R., Porcelli, L., Gristina, R., Favia, P., Fracassi, F., Trizio, I., Silvestris, N., Guida, G., Tommasi, S. and Sardella, E. (2019) Plasma-activated medium triggers cell death and the presentation of immune activating danger signals in melanoma and pancreatic cancer cells. Sci. Rep. 9, 4099.
    Pubmed KoreaMed CrossRef
  15. Bauer, G., Sersenová, D., Graves, D. B. and Machala, Z. (2019) Cold atmospheric plasma and plasma-activated medium trigger RONS-based tumor cell apoptosis. Sci. Rep. 9, 14210.
    Pubmed KoreaMed CrossRef
  16. Bavelloni, A., Piazzi, M., Raffini, M., Faenza, I. and Blalock, W. L. (2015) Prohibitin 2: at a communications crossroads. IUBMB Life 67, 239-254.
    Pubmed CrossRef
  17. Bekeschus, S., Rodder, K., Fregin, B., Otto, O., Lippert, M., Weltmann, K. D., Wende, K., Schmidt, A. and Gandhirajan, R. K. (2017) Toxicity and immunogenicity in murine melanoma following exposure to physical plasma-derived oxidants. Oxid. Med. Cell. Longev. 2017, 4396467.
    Pubmed KoreaMed CrossRef
  18. Bekeschus, S., Wende, K., Hefny, M. M., Rödder, K., Jablonowski, H., Schmidt, A., Woedtke, T., Weltmann, K.-D. and Benedikt, J. (2017) Oxygen atoms are critical in rendering THP-1 leukaemia cells susceptible to cold physical plasma-induced apoptosis. Sci. Rep. 7, 2791.
    Pubmed KoreaMed CrossRef
  19. Bernhardt, T., Semmler, M. L., Schafer, M., Bekeschus, S., Emmert, S. and Boeckmann, L. (2019) Plasma medicine: applications of cold atmospheric pressure plasma in dermatology. Oxid. Med. Cell. Longev. 2019, 3873928.
    Pubmed KoreaMed CrossRef
  20. Binenbaum, Y., Ben-David, G., Gil, Z., Slutsker, Y. Z., Ryzhkov, M. A., Felsteiner, J., Krasik, Y. E. and Cohen, J. T. (2017) Cold atmospheric plasma, created at the tip of an elongated flexible capillary using low electric current, can slow the progression of melanoma. PLoS One 12, e0169457.
    Pubmed KoreaMed CrossRef
  21. Breathnach, R., McDonnell, K. A., Chebbi, A., Callanan, J. J. and Dowling, D. P. (2018) Evaluation of the effectiveness of kINPen Med plasma jet and bioactive agent therapy in a rat model of wound healing. Biointerphases 13, 051002.
    Pubmed CrossRef
  22. Bunz, O., Mese, K., Funk, C., Wulf, M., Bailer, S. M., Piwowarczyk, A. and Ehrhardt, A. (2020) Cold atmospheric plasma as antiviral therapy - effect on human herpes simplex virus type 1. J.. Gen. Virol. 101, 208-215.
    Pubmed KoreaMed CrossRef
  23. Busco, G., Robert, E., Chettouh-Hammas, N., Pouvesle, J. M. and Grillon, C. (2020) The emerging potential of cold atmospheric plasma in skin biology. Free Radic. Biol. Med. 161, 290-304.
    Pubmed CrossRef
  24. Canady, J. (2020) Businesswire: Canady Helios Cold Plasma Scalpel Successfully Used by Chaim Sheba Medical Center Surgeons to Remove Inoperable Retroperitoneal Cancer. US Medical Innovations, LLC. Available from: https://www.businesswire.com/news/home/20200129005667/en/Canady-Helios-Cold-Plasma-Scalpel-Successfully-Chaim/ [accessed 2022 Nov 25].
  25. Chang, J. W., Kang, S. U., Shin, Y. S., Seo, S. J., Kim, Y. S., Yang, S. S., Lee, J.-S., Moon, E., Lee, K. and Kim, C.-H. (2015) Combination of NTP with cetuximab inhibited invasion/migration of cetuximab-resistant OSCC cells: involvement of NF-κB signaling. Sci. Rep. 5, 18208.
    Pubmed KoreaMed CrossRef
  26. Chauvin, J., Judee, F., Merbahi, N. and Vicendo, P. (2018) Effects of plasma activated medium on head and neck FaDu cancerous cells: comparison of 3D and 2D response. Anticancer Agents Med. Chem. 18, 776-783.
    Pubmed CrossRef
  27. Chen, G., Chen, Z., Wang, Z., Obenchain, R., Wen, D., Li, H., Wirz, R. E. and Gu, Z. (2021) Portable air-fed cold atmospheric plasma device for postsurgical cancer treatment. Sci. Adv. 7, eabg5686.
    Pubmed KoreaMed CrossRef
  28. Chen, Q., Sun, T., Wang, G., Zhang, M., Zhu, Y., Shi, X. and Ding, Z. (2022) Cuproptosis-related LncRNA signature for predicting prognosis of hepatocellular carcinoma: a comprehensive analysis. Dis. Markers 2022, 3265212.
    Pubmed KoreaMed CrossRef
  29. Chen, Z., Simonyan, H., Cheng, X., Gjika, E., Lin, L., Canady, J., Sherman, J. H., Young, C. and Keidar, M. (2017) A novel micro cold atmospheric plasma device for glioblastoma both in vitro and in vivo. Cancers (Basel) 9, 61.
    Pubmed KoreaMed CrossRef
  30. Cheng, X., Murphy, W., Recek, N., Yan, D., Cvelbar, U., Vesel, A., Mozetič, M., Canady, J., Keidar, M. and Sherman, J. H. (2014a) Synergistic effect of gold nanoparticles and cold plasma on glioblastoma cancer therapy. J. Phys. D: Appl. Phys. 47, 335402.
    CrossRef
  31. Cheng, X., Rajjoub, K., Shashurin, A., Yan, D., Sherman, J. H., Bian, K., Murad, F. and Keidar, M. (2017) Enhancing cold atmospheric plasma treatment of cancer cells by static magnetic field. Bioelectromagnetics 38, 53-62.
    Pubmed CrossRef
  32. Cheng, X., Sherman, J., Murphy, W., Ratovitski, E., Canady, J. and Keidar, M. (2014b) The effect of tuning cold plasma composition on glioblastoma cell viability. PLoS One 9, e98652.
    Pubmed KoreaMed CrossRef
  33. Choi, J.-S., Kim, J., Hong, Y.-J., Bae, W.-Y., Choi, E. H., Jeong, J.-W. and Park, H.-K. (2017) Evaluation of non-thermal plasma-induced anticancer effects on human colon cancer cells. Biomed. Opt. Express 8, 2649-2659.
    Pubmed KoreaMed CrossRef
  34. Conway, G. E., Casey, A., Milosavljevic, V., Liu, Y., Howe, O., Cullen, P. J. and Curtin, J. F. (2016) Non-thermal atmospheric plasma induces ROS-independent cell death in U373MG glioma cells and augments the cytotoxicity of temozolomide. Br. J. Cancer 114, 435-443.
    Pubmed KoreaMed CrossRef
  35. Conway, G. E., He, Z., Hutanu, A. L., Cribaro, G. P., Manaloto, E., Casey, A., Traynor, D., Milosavljevic, V., Howe, O., Barcia, C., Murray, J. T., Cullen, P. J. and Curtin, J. F. (2019) Cold Atmospheric Plasma induces accumulation of lysosomes and caspase-independent cell death in U373MG glioblastoma multiforme cells. Sci. Rep. 9, 12891.
    Pubmed KoreaMed CrossRef
  36. Corbella, C., Portal, S., Lin, L. and Keidar, M. (2021) Non-thermal plasma multi-jet platform based on a flexible matrix. Rev. Sci. Instrum. 92, 083505.
    Pubmed CrossRef
  37. Dai, X., Bazaka, K., Richard, D. J., Thompson, E. R. W. and Ostrikov, K. K. (2018) The emerging role of gas plasma in oncotherapy. Trends Biotechnol. 36, 1183-1198.
    Pubmed CrossRef
  38. Dai, X., Bazaka, K., Thompson, E. W. and Ostrikov, K. K. (2020a) Cold atmospheric plasma: a promising controller of cancer cell states. Cancers (Basel) 12, 3360.
    Pubmed KoreaMed CrossRef
  39. Dai, X., Cai, D., Wang, P., Nan, N., Yu, L., Zhang, Z., Zhou, R., Hua, D., Zhang, J., Ostrikov, K. K. and Thompson, E. (2022) Cold atmospheric plasmas target breast cancer stemness via modulating AQP3-19Y mediated AQP3-5K and FOXO1 K48-ubiquitination. Int. J. Biol. Sci. 18, 3544-3561.
    Pubmed KoreaMed CrossRef
  40. Dai, X., Luo, Y., Xu, Y. and Zhang, J. (2019) Key indexes and the emerging tool for tumor microenvironment editing. Am. J. Cancer Res. 9, 1027-1042.
    Pubmed KoreaMed
  41. Dai, X., Shen, L. and Zhang, J. (2023) Cold atmospheric plasma: redox homeostasis to treat cancers?. Trends Biotechnol. 41, 15-18.
    Pubmed CrossRef
  42. Dai, X., Wang, D. and Zhang, J. (2021) Programmed cell death, redox imbalance, and cancer therapeutics. Apoptosis 26, 385-414.
    Pubmed CrossRef
  43. Dai, X., Xiang, L., Li, T. and Bai, Z. (2016) Cancer hallmarks, biomarkers and breast cancer molecular subtypes. J. Cancer 7, 1281-1294.
    Pubmed KoreaMed CrossRef
  44. Dai, X. F., Zhang, Z. F., Zhang, J. Y. and Ostrikov, K. K. (2020b) Dosing: the key to precision plasma oncology. Plasma Processes Polym. 17, 1900178.
    CrossRef
  45. Davalli, P., Marverti, G., Lauriola, A. and D'Arca, D. (2018) Targeting oxidatively induced DNA damage response in cancer: opportunities for novel cancer therapies. Oxid. Med.Cell. Longev. 2018, 2389523.
    Pubmed KoreaMed CrossRef
  46. Dezest, M., Chavatte, L., Bourdens, M., Quinton, D., Camus, M., Garrigues, L., Descargues, P., Arbault, S., Burlet-Schiltz, O., Casteilla, L., Clement, F., Planat, V. and Bulteau, A. L. (2017) Mechanistic insights into the impact of Cold Atmospheric Pressure Plasma on human epithelial cell lines. Sci. Rep. 7, 41163.
    Pubmed KoreaMed CrossRef
  47. Duchesne, C., Frescaline, N., Blaise, O., Lataillade, J. J., Banzet, S., Dussurget, O. and Rousseau, A. (2021) Cold atmospheric plasma promotes killing of Staphylococcus aureus by macrophages. mSphere 6, e0021721.
    Pubmed KoreaMed CrossRef
  48. Ermakov, A. M., Ermakova, O. N., Afanasyeva, V. A. and Popov, A. L. (2021) Dose-dependent effects of cold atmospheric argon plasma on the mesenchymal stem and osteosarcoma cells in vitro. Int. J. Mol. Sci. 22, 6797.
    Pubmed KoreaMed CrossRef
  49. Estarabadi, H., Atyabi, S. A., Tavakkoli, S., Noormohammadi, Z., Gholami, M. R., Ghiaseddin, A. and Irani, S. (2021) Cold atmospheric plasma induced genotoxicity and cytotoxicity in esophageal cancer cells. Mol. Biol. Rep. 48, 1323-1333.
    Pubmed CrossRef
  50. Eto, K., Ishinada, C., Suemoto, T., Hyakutake, K., Tanaka, H. and Hori, M. (2021) Differential data on the responsiveness of multiple cell types to cell death induced by non-thermal atmospheric pressure plasma-activated solutions. Data Brief 36, 106995.
    Pubmed KoreaMed CrossRef
  51. Feil, L., Koch, A., Utz, R., Ackermann, M., Barz, J., Stope, M., Krämer, B., Wallwiener, D., Brucker, S. Y. and Weiss, M. (2020) Cancer-selective treatment of cancerous and non-cancerous human cervical cell models by a non-thermally operated electrosurgical argon plasma device. Cancers (Basel) 12, 1037.
    Pubmed KoreaMed CrossRef
  52. Friedman, P. C., Miller, V., Fridman, G., Lin, A. and Fridman, A. (2017) Successful treatment of actinic keratoses using nonthermal atmospheric pressure plasma: a case series. J. Am. Acad. Dermatol. 76, 349-350.
    Pubmed CrossRef
  53. Gangemi, S., Petrarca, C., Tonacci, A., Di Gioacchino, M., Musolino, C. and Allegra, A. (2022) Cold atmospheric plasma targeting hematological malignancies: potentials and problems of clinical translation. Antioxidants (Basel) 11, 1592.
    Pubmed KoreaMed CrossRef
  54. Gay-Mimbrera, J., García, M. C., Isla-Tejera, B., Rodero-Serrano, A., García-Nieto, A. V. and Ruano, J. (2016) Clinical and biological principles of cold atmospheric plasma application in skin cancer. Adv. Ther. 33, 894-909.
    Pubmed KoreaMed CrossRef
  55. Gelbrich, N., Miebach, L., Berner, J., Freund, E., Saadati, F., Schmidt, A., Stope, M., Zimmermann, U., Burchardt, M. and Bekeschus, S. (2023) Medical gas plasma augments bladder cancer cell toxicity in preclinical models and patient-derived tumor tissues. J. Adv. Res. 47, 209-223.
    Pubmed KoreaMed CrossRef
  56. Gjika, E., Pal-Ghosh, S., Kirschner, M. E., Lin, L., Sherman, J. H., Stepp, M. A. and Keidar, M. (2020) Combination therapy of cold atmospheric plasma (CAP) with temozolomide in the treatment of U87MG glioblastoma cells. Sci. Rep. 10, 16495.
    Pubmed KoreaMed CrossRef
  57. Golpour, M., Alimohammadi, M., Sohbatzadeh, F., Fattahi, S., Bekeschus, S. and Rafiei, A. (2022) Cold atmospheric pressure plasma treatment combined with starvation increases autophagy and apoptosis in melanoma in vitro and in vivo. Exp.. Dermatol. 31, 1016-1028.
    Pubmed CrossRef
  58. Golubitskaya, E. A., Troitskaya, O. S., Yelak, E. V., Gugin, P. P., Richter, V. A., Schweigert, I. V., Zakrevsky, D. E. and Koval, O. A. (2019) Cold physical plasma decreases the viability of lung adenocarcinoma cells. Acta Naturae 11, 16-19.
    Pubmed KoreaMed CrossRef
  59. Gorrini, C., Harris, I. S. and Mak, T. W. (2013) Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 12, 931-947.
    Pubmed CrossRef
  60. Griseti, E., Kolosnjaj-Tabi, J., Gibot, L., Fourquaux, I., Rols, M.-P., Yousfi, M., Merbahi, N. and Golzio, M. (2019) Pulsed electric field treatment enhances the cytotoxicity of plasma-activated liquids in a three-dimensional human colorectal cancer cell model. Sci. Rep. 9, 7583.
    Pubmed KoreaMed CrossRef
  61. Griseti, E., Merbahi, N. and Golzio, M. (2020) Anti-cancer potential of two plasma-activated liquids: implication of long-lived reactive oxygen and nitrogen species. Cancers (Basel) 12, 721.
    Pubmed KoreaMed CrossRef
  62. Guerrero-Preston, R., Ogawa, T., Uemura, M., Shumulinsky, G., Valle, B. L., Pirini, F., Ravi, R., Sidransky, D., Keidar, M. and Trink, B. (2014) Cold atmospheric plasma treatment selectively targets head and neck squamous cell carcinoma cells. Int. J. Mol. Med. 34, 941-946.
    Pubmed KoreaMed CrossRef
  63. Gümbel, D., Gelbrich, N., Napp, M., Daeschlein, G., Kramer, A., Sckell, A., Burchardt, M., Ekkernkamp, A. and Stope, M. B. (2017a) Peroxiredoxin expression of human osteosarcoma cells is influenced by cold atmospheric plasma treatment. Anticancer Res. 37, 1031-1038.
    CrossRef
  64. Gümbel, D., Gelbrich, N., Weiss, M., Napp, M., Daeschlein, G., Sckell, A., Ender, S. A., Kramer, A., Burchardt, M., Ekkernkamp, A. and Stope, M. B. (2016) New treatment options for osteosarcoma - inactivation of osteosarcoma cells by cold atmospheric plasma. Anticancer Res. 36, 5915-5922.
    Pubmed CrossRef
  65. Gümbel, D., Suchy, B., Wien, L., Gelbrich, N., Napp, M., Kramer, A., Ekkernkamp, A., Daeschlein, G. and Stope, M. B. (2017b) Comparison of cold atmospheric plasma devices' efficacy on osteosarcoma and fibroblastic in vitro cell models. Anticancer Res. 37, 5407-5414.
  66. Gunes, S., He, Z., Tsoukou, E., Ng, S. W., Boehm, D., Pinheiro Lopes, B., Bourke, P., Malone, R., Cullen, P. J., Wang, W. and Curtin, J. (2022) Cell death induced in glioblastoma cells by plasma-activated-liquids (PAL) is primarily mediated by membrane lipid peroxidation and not ROS influx. PLoS One 17, e0274524.
    Pubmed KoreaMed CrossRef
  67. Guo, B., Li, W., Liu, Y., Xu, D., Liu, Z. and Huang, C. (2022) Aberrant expressional profiling of small RNA by cold atmospheric plasma treatment in human chronic myeloid leukemia cells. Front. Genet. 12, 809658.
    Pubmed KoreaMed CrossRef
  68. Guo, B., Pomicter, A. D., Li, F., Bhatt, S., Chen, C., Li, W., Qi, M., Huang, C., Deininger, M. W., Kong, M. G. and Chen, H.-L. (2021) Trident cold atmospheric plasma blocks three cancer survival pathways to overcome therapy resistance. Proc. Natl.. Acad. Sci. U. S. A. 118, e2107220118.
    Pubmed KoreaMed CrossRef
  69. Ha, J.-H. and Kim, Y.-J. (2021) Photodynamic and cold atmospheric plasma combination therapy using polymeric nanoparticles for the synergistic treatment of cervical cancer. Int. J.. Mol. Sci. 22, 1172.
    Pubmed KoreaMed CrossRef
  70. Haertel, B., von Woedtke, T., Weltmann, K. D. and Lindequist, U. (2014) Non-thermal atmospheric-pressure plasma possible application in wound healing. Biomol. Ther. (Seoul) 22, 477-490.
    Pubmed KoreaMed CrossRef
  71. Hamouda, I., Labay, C., Cvelbar, U., Ginebra, M.-P. and Canal, C. (2021) Selectivity of direct plasma treatment and plasma-conditioned media in bone cancer cell lines. Sci. Rep. 11, 17521.
    Pubmed KoreaMed CrossRef
  72. Han, I., Choi, S. A., Kim, S. I., Choi, E. H., Lee, Y. J. and Kim, Y. (2021) Improvement of cell growth of uterosacral ligament fibroblast derived from pelvic organ prolapse patients by cold atmospheric plasma treated liquid. Cells 10, 2728.
    Pubmed KoreaMed CrossRef
  73. Hara, H., Taniguchi, M., Kobayashi, M., Kamiya, T. and Adachi, T. (2015) Plasma-activated medium-induced intracellular zinc liberation causes death of SH-SY5Y cells. Arch Biochem. Biophys. 584, 51-60.
    Pubmed CrossRef
  74. Haralambiev, L., Nitsch, A., Einenkel, R., Muzzio, D. O., Gelbrich, N., Burchardt, M., Zygmunt, M., Ekkernkamp, A., Stope, M. B. and Gümbel, D. (2020a) The effect of cold atmospheric plasma on the membrane permeability of human osteosarcoma cells. Anticancer Res. 40, 841-846.
    Pubmed CrossRef
  75. Haralambiev, L., Nitsch, A., Jacoby, J. M., Strakeljahn, S., Bekeschus, S., Mustea, A., Ekkernkamp, A. and Stope, M. B. (2020b) Cold atmospheric plasma treatment of chondrosarcoma cells affects proliferation and cell membrane permeability. Int. J. Mol. Sci. 21, 2291.
    Pubmed KoreaMed CrossRef
  76. Haralambiev, L., Wien, L., Gelbrich, N., Kramer, A., Mustea, A., Burchardt, M., Ekkernkamp, A., Stope, M. B. and Gümbel, D. (2019) Effects of cold atmospheric plasma on the expression of chemokines, growth factors, TNF superfamily members, interleukins, and cytokines in human osteosarcoma cells. Anticancer Res. 39, 151-157.
    Pubmed CrossRef
  77. Haralambiev, L., Wien, L., Gelbrich, N., Lange, J., Bakir, S., Kramer, A., Burchardt, M., Ekkernkamp, A., Gümbel, D. and Stope, M. B. (2020c) Cold atmospheric plasma inhibits the growth of osteosarcoma cells by inducing apoptosis, independent of the device used. Oncol. Lett. 19, 283-290.
    Pubmed KoreaMed CrossRef
  78. Hattori, N., Yamada, S., Torii, K., Takeda, S., Nakamura, K., Tanaka, H., Kajiyama, H., Kanda, M., Fujii, T., Nakayama, G., Sugimoto, H., Koike, M., Nomoto, S., Fujiwara, M., Mizuno, M., Hori, M. and Kodera, Y. (2015) Effectiveness of plasma treatment on pancreatic cancer cells. Int. J. Oncol. 47, 1655-1662.
    Pubmed KoreaMed CrossRef
  79. He, Z., Charleton, C., Devine, R. W., Kelada, M., Walsh, J. M. D., Conway, G. E., Gunes, S., Mondala, J. R. M., Tian, F., Tiwari, B., Kinsella, G. K., Malone, R., O'Shea, D., Devereux, M., Wang, W., Cullen, P. J., Stephens, J. C. and Curtin, J. F. (2021) Enhanced pyrazolopyrimidinones cytotoxicity against glioblastoma cells activated by ROS-Generating cold atmospheric plasma. Eur. J. Med. Chem. 224, 113736.
    Pubmed CrossRef
  80. He, Z., Liu, K., Manaloto, E., Casey, A., Cribaro, G. P., Byrne, H. J., Tian, F., Barcia, C., Conway, G. E., Cullen, P. J. and Curtin, J. F. (2018) Cold atmospheric plasma induces ATP-dependent endocytosis of nanoparticles and synergistic U373MG cancer cell death. Sci. Rep. 8, 5298.
    Pubmed KoreaMed CrossRef
  81. He, Z., Liu, K., Scally, L., Manaloto, E., Gunes, S., Ng, S. W., Maher, M., Tiwari, B., Byrne, H. J., Bourke, P., Tian, F., Cullen, P. J. and Curtin, J. F. (2020) Cold atmospheric plasma stimulates clathrin-dependent endocytosis to repair oxidised membrane and enhance uptake of nanomaterial in glioblastoma multiforme cells. Sci. Rep. 10, 6985.
    Pubmed KoreaMed CrossRef
  82. Hirst, A. M., Simms, M. S., Mann, V. M., Maitland, N. J., O'Connell, D. and Frame, F. M. (2015) Low-temperature plasma treatment induces DNA damage leading to necrotic cell death in primary prostate epithelial cells. Br. J. Cancer 112, 1536-1545.
    Pubmed KoreaMed CrossRef
  83. Hoffmann, C., Berganza, C. and Zhang, J. (2013) Cold Atmospheric Plasma: methods of production and application in dentistry and oncology. Med. Gas Res. 3, 21.
    Pubmed KoreaMed CrossRef
  84. Hong, Y. F., Kang, J. G., Lee, H. Y., Uhm, H. S., Moon, E. and Park, Y. H. (2009) Sterilization effect of atmospheric plasma on Escherichia coli and Bacillus subtilis endospores. Lett. Appl. Microbiol. 48, 33-37.
    Pubmed CrossRef
  85. Hua, D., Cai, D., Ning, M., Yu, L., Zhang, Z., Han, P. and Dai, X. (2021) Cold atmospheric plasma selectively induces G0/G1 cell cycle arrest and apoptosis in AR-independent prostate cancer cells. J. Cancer 12, 5977-5986.
    Pubmed KoreaMed CrossRef
  86. Huang, J., Zhang, J., Wang, F., Zhang, B. and Tang, X. (2022) Comprehensive analysis of cuproptosis-related genes in immune infiltration and diagnosis in ulcerative colitis. Front. Immunol. 13, 1008146.
    Pubmed KoreaMed CrossRef
  87. Irani, S., Shahmirani, Z., Atyabi, S. M. and Mirpoor, S. (2015) Induction of growth arrest in colorectal cancer cells by cold plasma and gold nanoparticles. Arch. Med. Sci. 11, 1286-1295.
    Pubmed KoreaMed CrossRef
  88. Isbary, G., Morfill, G., Schmidt, H. U., Georgi, M., Ramrath, K., Heinlin, J., Karrer, S., Landthaler, M., Shimizu, T., Steffes, B., Bunk, W., Monetti, R., Zimmermann, J. L., Pompl, R. and Stolz, W. (2010) A first prospective randomized controlled trial to decrease bacterial load using cold atmospheric argon plasma on chronic wounds in patients. Br. J.. Dermatol. 163, 78-82.
    Pubmed CrossRef
  89. Isbary, G., Shimizu, T., Li, Y. F., Stolz, W., Thomas, H. M., Morfill, G. E. and Zimmermann, J. L. (2013) Cold atmospheric plasma devices for medical issues. Expert Rev. Med. Devices 10, 367-377.
    Pubmed CrossRef
  90. Ishaq, M., Evans, M. D. M. and Ostrikov, K. K. (2014) Atmospheric pressure gas plasma-induced colorectal cancer cell death is mediated by Nox2-ASK1 apoptosis pathways and oxidative stress is mitigated by Srx-Nrf2 anti-oxidant system. Biochim. Biophys. Acta 1843, 2827-2837.
    Pubmed CrossRef
  91. Jacoby, J. M., Strakeljahn, S., Nitsch, A., Bekeschus, S., Hinz, P., Mustea, A., Ekkernkamp, A., Tzvetkov, M. V., Haralambiev, L. and Stope, M. B. (2020) An innovative therapeutic option for the treatment of skeletal sarcomas: elimination of osteo- and Ewing's sarcoma cells using physical gas plasma. Int. J. Mol. Sci. 21, 4460.
    Pubmed KoreaMed CrossRef
  92. Jalili, A., Irani, S. and Mirfakhraie, R. (2016) Combination of cold atmospheric plasma and iron nanoparticles in breast cancer: gene expression and apoptosis study. Onco Targets Ther. 9, 5911-5917.
    Pubmed KoreaMed CrossRef
  93. Ji, H. W., Kim, H., Kim, H. W., Yun, S. H., Park, J. E., Choi, E. H. and Kim, S. J. (2020) Genome-wide comparison of the target genes of the reactive oxygen species and non-reactive oxygen species constituents of cold atmospheric plasma in cancer cells. Cancers (Basel) 12, 2640.
    Pubmed KoreaMed CrossRef
  94. Jo, A., Bae, J. H., Yoon, Y. J., Chung, T. H., Lee, E.-W., Kim, Y.-H., Joh, H. M. and Chung, J. W. (2022a) Plasma-activated medium induces ferroptosis by depleting FSP1 in human lung cancer cells. Cell Death Dis. 13, 212.
    Pubmed KoreaMed CrossRef
  95. Jo, A., Joh, H. M., Bae, J. H., Kim, S. J., Chung, T. H. and Chung, J. W. (2022b) Plasma activated medium prepared by a bipolar microsecond-pulsed atmospheric pressure plasma jet array induces mitochondria-mediated apoptosis in human cervical cancer cells. PLoS One 17, e0272805.
    Pubmed KoreaMed CrossRef
  96. Joerger, A. C. and Fersht, A. R. (2008) Structural biology of the tumor suppressor p53. Annu. Rev. Biochem. 77, 557-582.
    Pubmed CrossRef
  97. Jones, O., Cheng, X., Murthy, S. R. K., Ly, L., Zhuang, T., Basadonna, G., Keidar, M. and Canady, J. (2021) The synergistic effect of Canady Helios cold atmospheric plasma and a FOLFIRINOX regimen for the treatment of cholangiocarcinoma in vitro. Sci. Rep. 11, 8967.
    Pubmed KoreaMed CrossRef
  98. Kang, S. U., Cho, J. H., Chang, J. W., Shin, Y. S., Kim, K. I., Park, J. K., Yang, S. S., Lee, J. S., Moon, E., Lee, K. and Kim, C. H. (2014) Nonthermal plasma induces head and neck cancer cell death: the potential involvement of mitogen-activated protein kinase-dependent mitochondrial reactive oxygen species. Cell Death Dis. 5, e1056.
    Pubmed KoreaMed CrossRef
  99. Kaushik, N., Lee, S. J., Choi, T. G., Baik, K. Y., Uhm, H. S., Kim, C. H., Kaushik, N. K. and Choi, E. H. (2015) Non-thermal plasma with 2-deoxy-D-glucose synergistically induces cell death by targeting glycolysis in blood cancer cells. Sci. Rep. 5, 8726.
    Pubmed KoreaMed CrossRef
  100. Kaushik, N. K., Attri, P., Kaushik, N. and Choi, E. H. (2013) A preliminary study of the effect of DBD plasma and osmolytes on T98G brain cancer and HEK non-malignant cells. Molecules 18, 4917-4928.
    Pubmed KoreaMed CrossRef
  101. Kaushik, N. K., Ghimire, B., Li, Y., Adhikari, M., Veerana, M., Kaushik, N., Jha, N., Adhikari, B., Lee, S. J., Masur, K., von Woedtke, T., Weltmann, K. D. and Choi, E. H. (2018) Biological and medical applications of plasma-activated media, water and solutions. Biol. Chem. 400, 39-62.
    Pubmed CrossRef
  102. Kaushik, N. K., Kaushik, N., Adhikari, M., Ghimire, B., Linh, N. N., Mishra, Y. K., Lee, S. J. and Choi, E. H. (2019) Preventing the solid cancer progression via release of anticancer-cytokines in co-culture with cold plasma-stimulated macrophages. Cancers (Basel) 11, 842.
    Pubmed KoreaMed CrossRef
  103. Keidar, M., Walk, R., Shashurin, A., Srinivasan, P., Sandler, A., Dasgupta, S., Ravi, R., Guerrero-Preston, R. and Trink, B. (2011) Cold plasma selectivity and the possibility of a paradigm shift in cancer therapy. Br. J. Cancer 105, 1295-1301.
    Pubmed KoreaMed CrossRef
  104. Keidar, M., Yan, D., Beilis, I. I., Trink, B. and Sherman, J. H. (2018) Plasmas for treating cancer: opportunities for adaptive and self-adaptive approaches. Trends Biotechnol. 36, 586-593.
    Pubmed CrossRef
  105. Kenari, A. J., Siadati, S. N., Abedian, Z., Sohbatzadeh, F., Amiri, M., Gorji, K. E., Babapour, H., Zabihi, E., Ghoreishi, S. M., Mehraeen, R. and Monfared, A. S. (2021) Therapeutic effect of cold atmospheric plasma and its combination with radiation as a novel approach on inhibiting cervical cancer cell growth (HeLa cells). Bioorg. Chem. 111, 104892.
    Pubmed CrossRef
  106. Khabipov, A., Freund, E., Liedtke, K. R., Käding, A., Riese, J., van der Linde, J., Kersting, S., Partecke, L.-I. and Bekeschus, S. (2021) Murine macrophages modulate their inflammatory profile in response to gas plasma-inactivated pancreatic cancer cells. Cancers (Basel) 13, 2525.
    Pubmed KoreaMed CrossRef
  107. Kim, C.-H., Bahn, J. H., Lee, S.-H., Kim, G.-Y., Jun, S.-I., Lee, K. and Baek, S. J. (2010a) Induction of cell growth arrest by atmospheric non-thermal plasma in colorectal cancer cells. J. Biotechnol. 150, 530-538.
    Pubmed CrossRef
  108. Kim, C.-H., Kwon, S., Bahn, J. H., Lee, K., Jun, S. I., Rack, P. D. and Baek, S. J. (2010b) Effects of atmospheric nonthermal plasma on invasion of colorectal cancer cells. Appl. Phys. Lett. 96, 243701.
    Pubmed KoreaMed CrossRef
  109. Kim, G. C., Kim, G. J., Park, S. R., Jeon, S. M., Seo, H. J., Iza, F. and Lee, J. K. (2009) Air plasma coupled with antibody-conjugated nanoparticles: a new weapon against cancer. J. Phys. D: Appl. Phys. 42, 032005.
    CrossRef
  110. Kim, H.-Y., Agrahari, G., Lee, M. J., Tak, L.-J., Ham, W.-K. and Kim, T.-Y. (2021) Low-temperature argon plasma regulates skin moisturizing and melanogenesis-regulating markers through yes-associated protein. Int. J. Mol. Sci. 22, 1895.
    Pubmed KoreaMed CrossRef
  111. Kim, H. W., Jeong, D., Ham, J., Kim, H., Ji, H. W., Choi, E. H. and Kim, S. J. (2020) ZNRD1 and its antisense long noncoding RNA ZNRD1-AS1 are oppositely regulated by cold atmospheric plasma in breast cancer cells. Oxid. Med. Cell. Longev. 2020, 9490567.
    Pubmed KoreaMed CrossRef
  112. Kim, J. Y., Ballato, J., Foy, P., Hawkins, T., Wei, Y., Li, J. and Kim, S.-O. (2011) Apoptosis of lung carcinoma cells induced by a flexible optical fiber-based cold microplasma. Biosens. Bioelectron. 28, 333-338.
    Pubmed CrossRef
  113. Kim, S.-J., Seong, M.-J., Mun, J.-J., Bae, J.-H., Joh, H.-M. and Chung, T.-H. (2022) Differential sensitivity of melanoma cells and their non-cancerous counterpart to cold atmospheric plasma-induced reactive oxygen and nitrogen species. Int. J. Mol. Sci. 23, 14092.
    Pubmed KoreaMed CrossRef
  114. Kim, S.-Y., Kim, H.-J., Kang, S. U., Kim, Y. E., Park, J. K., Shin, Y. S., Kim, Y. S., Lee, K. and Kim, C.-H. (2015) Non-thermal plasma induces AKT degradation through turn-on the MUL1 E3 ligase in head and neck cancer. Oncotarget 6, 33382-33396.
    Pubmed KoreaMed CrossRef
  115. Kletschkus, K., Haralambiev, L., Nitsch, A., Pfister, F., Klinkmann, G., Kramer, A., Bekeschus, S., Mustea, A. and Stope, M. B. (2020) The application of a low-temperature physical plasma device operating under atmospheric pressure leads to the production of toxic NO2. Anticancer Res. 40, 2591-2599.
    Pubmed CrossRef
  116. Koensgen, D., Besic, I., Gümbel, D., Kaul, A., Weiss, M., Diesing, K., Kramer, A., Bekeschus, S., Mustea, A. and Stope, M. B. (2017) Cold atmospheric plasma (CAP) and CAP-stimulated cell culture media suppress ovarian cancer cell growth - a putative treatment option in ovarian cancer therapy. Anticancer Res. 37, 6739-6744.
    CrossRef
  117. Koinuma, H., Ohkubo, H., Hashimoto, T., Inomata, K., Shiraishi, T., Miyanaga, A. and Hayashi, S. (1992) Development and application of a microbeam plasma generator. Appl. Phys. Lett. 60, 816-817.
    CrossRef
  118. Kordt, M., Trautmann, I., Schlie, C., Lindner, T., Stenzel, J., Schildt, A., Boeckmann, L., Bekeschus, S., Kurth, J., Krause, B. J., Vollmar, B. and Grambow, E. (2021) Multimodal imaging techniques to evaluate the anticancer effect of cold atmospheric pressure plasma. Cancers (Basel) 13, 2483.
    Pubmed KoreaMed CrossRef
  119. Köritzer, J., Boxhammer, V., Schäfer, A., Shimizu, T., Klämpfl, T. G., Li, Y.-F., Welz, C., Schwenk-Zieger, S., Morfill, G. E., Zimmermann, J. L. and Schlegel, J. (2013) Restoration of sensitivity in chemo-resistant glioma cells by cold atmospheric plasma. PLoS One 8, e64498.
    Pubmed KoreaMed CrossRef
  120. Kugler, P., Becker, S., Welz, C., Wiesmann, N., Sax, J., Buhr, C. R., Thoma, M. H., Brieger, J. and Eckrich, J. (2022) Cold atmospheric plasma reduces vessel density and increases vascular permeability and apoptotic cell death in solid tumors. Cancers (Basel) 14, 2432.
    Pubmed KoreaMed CrossRef
  121. Kurake, N., Ishikawa, K., Tanaka, H., Hashizume, H., Nakamura, K., Kajiyama, H., Toyokuni, S., Kikkawa, F., Mizuno, M. and Hori, M. (2019) Non-thermal plasma-activated medium modified metabolomic profiles in the glycolysis of U251SP glioblastoma. Arch. Biochem. Biophys. 662, 83-92.
    Pubmed CrossRef
  122. Kurake, N., Tanaka, H., Ishikawa, K., Kondo, T., Sekine, M., Nakamura, K., Kajiyama, H., Kikkawa, F., Mizuno, M. and Hori, M. (2016) Cell survival of glioblastoma grown in medium containing hydrogen peroxide and/or nitrite, or in plasma-activated medium. Arch. Biochem. Biophys. 605, 102-108.
    Pubmed CrossRef
  123. Kurita, H., Haruta, N., Uchihashi, Y., Seto, T. and Takashima, K. (2020) Strand breaks and chemical modification of intracellular DNA induced by cold atmospheric pressure plasma irradiation. PLoS One 15, e0232724.
    Pubmed KoreaMed CrossRef
  124. Lackmann, J. W., Schneider, S., Edengeiser, E., Jarzina, F., Brinckmann, S., Steinborn, E., Havenith, M., Benedikt, J. and Bandow, J. E. (2013) Photons and particles emitted from cold atmospheric-pressure plasma inactivate bacteria and biomolecules independently and synergistically. J. R. Soc. Interface 10, 20130591.
    Pubmed KoreaMed CrossRef
  125. Laroussi, M. (2018) Plasma medicine: a brief introduction. Plasma 1, 47-60.
    CrossRef
  126. Laroussi, M. and Lu, X. (2005) Room-temperature atmospheric pressure plasma plume for biomedical applications. Appl. Phys. Lett. 87, 113902.
    CrossRef
  127. Laroussi, M., Tendero, C., Lu, X., Alla, S. and Hynes, W. L. (2006) Inactivation of bacteria by the plasma pencil. Plasma Processes Polym. 3, 470-473.
    CrossRef
  128. Lee, J., Moon, H., Ku, B., Lee, K., Hwang, C.-Y. and Baek, S. J. (2020) Anticancer effects of cold atmospheric plasma in canine osteosarcoma cells. Int. J. Mol. Sci. 21, 4556.
    Pubmed KoreaMed CrossRef
  129. Lee, S., Lee, H., Bae, H., Choi, E. H. and Kim, S. J. (2016) Epigenetic silencing of miR-19a-3p by cold atmospheric plasma contributes to proliferation inhibition of the MCF-7 breast cancer cell. Sci. Rep. 6, 30005.
    Pubmed KoreaMed CrossRef
  130. Lee, S., Lee, H., Jeong, D., Ham, J., Park, S., Choi, E. H. and Kim, S. J. (2017) Cold atmospheric plasma restores tamoxifen sensitivity in resistant MCF-7 breast cancer cell. Free Radic. Biol. Med. 110, 280-290.
    Pubmed CrossRef
  131. Lee, S., Park, S., Lee, H., Jeong, D., Ham, J., Choi, E. H. and Kim, S. J. (2018) ChIP-seq analysis reveals alteration of H3K4 trimethylation occupancy in cancer-related genes by cold atmospheric plasma. Free Radic. Biol. Med. 126, 133-141.
    Pubmed CrossRef
  132. Lehmann, S., Bien-Möller, S., Marx, S., Bekeschus, S., Schroeder, H. W. S., Mustea, A. and Stope, M. B. (2023) Devitalization of glioblastoma cancer cells by non-invasive physical plasma: modulation of proliferative signalling cascades. Anticancer Res. 43, 7-18.
    Pubmed CrossRef
  133. Li, W., Yu, H., Ding, D., Chen, Z., Wang, Y., Wang, S., Li, X., Keidar, M. and Zhang, W. (2019) Cold atmospheric plasma and iron oxide-based magnetic nanoparticles for synergetic lung cancer therapy. Free Radic. Biol. Med. 130, 71-81.
    Pubmed CrossRef
  134. Li, Y., Ho Kang, M., Sup Uhm, H., Joon Lee, G., Ha Choi, E. and Han, I. (2017) Effects of atmospheric-pressure non-thermal bio-compatible plasma and plasma activated nitric oxide water on cervical cancer cells. Sci. Rep. 7, 45781.
    Pubmed KoreaMed CrossRef
  135. Li, Y., Tang, T., Lee, H. and Song, K. (2021a) Cold atmospheric pressure plasma-activated medium induces selective cell death in human hepatocellular carcinoma cells independently of singlet oxygen, hydrogen peroxide, nitric oxide and nitrite/nitrate. Int. J. Mol. Sci. 22, 5548.
    Pubmed KoreaMed CrossRef
  136. Li, Y., Tang, T., Lee, H. J. and Song, K. (2021b) Selective anti-cancer effects of plasma-activated medium and its high efficacy with cisplatin on hepatocellular carcinoma with cancer stem cell characteristics. Int. J. Mol. Sci. 22, 3956.
    Pubmed KoreaMed CrossRef
  137. Lin, A., De Backer, J., Quatannens, D., Cuypers, B., Verswyvel, H., De La Hoz, E. C., Ribbens, B., Siozopoulou, V., Van Audenaerde, J., Marcq, E., Lardon, F., Laukens, K., Vanlanduit, S., Smits, E. and Bogaerts, A. (2022) The effect of local non-thermal plasma therapy on the cancer-immunity cycle in a melanoma mouse model. Bioeng. Transl. Med. 7, e10314.
    Pubmed KoreaMed CrossRef
  138. Lin, A., Gorbanev, Y., De Backer, J., Van Loenhout, J., Van Boxem, W., Lemiere, F., Cos, P., Dewilde, S., Smits, E. and Bogaerts, A. (2019) Non-thermal plasma as a unique delivery system of short-lived reactive oxygen and nitrogen species for immunogenic cell death in melanoma cells. Adv. Sci.. (Weinh.) 6, 1802062.
    Pubmed KoreaMed CrossRef
  139. Lin, A. G., Xiang, B., Merlino, D. J., Baybutt, T. R., Sahu, J., Fridman, A., Snook, A. E. and Miller, V. (2018) Non-thermal plasma induces immunogenic cell death in vivo in murine CT26 colorectal tumors. Oncoimmunology 7, e1484978.
    Pubmed KoreaMed CrossRef
  140. Liu, Z., Wang, L., Xing, Q., Liu, X., Hu, Y., Li, W., Yan, Q., Liu, R. and Huang, N. (2022) Identification of GLS as a cuproptosis-related diagnosis gene in acute myocardial infarction. Front. Cardiovasc. Med. 9, 1016081.
    Pubmed KoreaMed CrossRef
  141. Liu, Z., Zheng, Y., Dang, J., Zhang, J. and Zhang, J. (2019) A novel antifungal plasma-activated hydrogel. ACS Appl.. Mater. Interfaces 11, 22941-22949.
    Pubmed CrossRef
  142. Lupu, A.-R., Georgescu, N., Călugăru, A., Cremer, L., Szegli, G. and Kerek, F. (2009) The effects of cold atmospheric plasma jets on B16 and COLO320 tumoral cells. Roum.. Arch. Microbiol. Immunol. 68, 136-144.
  143. Ly, L., Cheng, X., Murthy, S. R. K., Jones, O. Z., Zhuang, T., Gitelis, S., Blank, A. T., Nissan, A., Adileh, M., Colman, M., Keidar, M., Basadonna, G. and Canady, J. (2022) Canady cold helios plasma reduces soft tissue sarcoma viability by inhibiting proliferation, disrupting cell cycle, and inducing apoptosis: a preliminary report. Molecules 27, 4168.
    Pubmed KoreaMed CrossRef
  144. Ma, Y., Ha, C. S., Hwang, S. W., Lee, H. J., Kim, G. C., Lee, K. W. and Song, K. (2014) Non-thermal atmospheric pressure plasma preferentially induces apoptosis in p53-mutated cancer cells by activating ROS stress-response pathways. PLoS One 9, e91947.
    Pubmed KoreaMed CrossRef
  145. Mahdikia, H., Shokri, B. and Majidzadeh-A, K. (2021) The feasibility study of plasma-activated water as a physical therapy to induce apoptosis in melanoma cancer cells in-vitro. Iran. J. Pharm. Res. 20, 337-350.
    Pubmed KoreaMed CrossRef
  146. Makinoshima, H., Takita, M., Matsumoto, S., Yagishita, A., Owada, S., Esumi, H. and Tsuchihara, K. (2014) Epidermal growth factor receptor (EGFR) signaling regulates global metabolic pathways in EGFR-mutated lung adenocarcinoma. J. Biol. Chem. 289, 20813-20823.
    Pubmed KoreaMed CrossRef
  147. Manaloto, E., Gowen, A. A., Lesniak, A., He, Z., Casey, A., Cullen, P. J. and Curtin, J. F. (2020) Cold atmospheric plasma induces silver nanoparticle uptake, oxidative dissolution and enhanced cytotoxicity in glioblastoma multiforme cells. Arch. Biochem. Biophys. 689, 108462.
    Pubmed CrossRef
  148. Mark, P. (2019) Plasma Scalpel Takes on Cancer: a New Tool Enters a Pivotal Pilot Study. Scientific American. Available from: https://www.scientificamerican.com/article/plasma-scalpel-takes-on-cancer/.
  149. Marzi, J., Stope, M. B., Henes, M., Koch, A., Wenzel, T., Holl, M., Layland, S. L., Neis, F., Bösmüller, H., Ruoff, F., Templin, M., Krämer, B., Staebler, A., Barz, J., Carvajal Berrio, D. A., Enderle, M., Loskill, P. M., Brucker, S. Y., Schenke-Layland, K. and Weiss, M. (2022) Noninvasive physical plasma as innovative and tissue-preserving therapy for women positive for cervical intraepithelial neoplasia. Cancers (Basel) 14, 1933.
    Pubmed KoreaMed CrossRef
  150. Mateu-Sanz, M., Ginebra, M.-P., Tornín, J. and Canal, C. (2022) Cold atmospheric plasma enhances doxorubicin selectivity in metastasic bone cancer. Free Radic. Biol. Med. 189, 32-41.
    Pubmed CrossRef
  151. Mateu-Sanz, M., Tornín, J., Brulin, B., Khlyustova, A., Ginebra, M.-P., Layrolle, P. and Canal, C. (2020) Cold plasma-treated Ringer's saline: a weapon to target osteosarcoma. Cancers (Basel) 12, 227.
    Pubmed KoreaMed CrossRef
  152. Metelmann, H.-R., Nedrelow, D. S., Seebauer, C., Schuster, M., von Woedtke, T., Weltmann, K.-D., Kindler, S., Metelmann, P. H., Finkelstein, S. E., Von Hoff, D. D. and Podmelle, F. (2015) Head and neck cancer treatment and physical plasma. Clin. Plasma Med. 3, 17-23.
    CrossRef
  153. Metelmann, H.-R., Seebauer, C., Miller, V., Fridman, A., Bauer, G., Graves, D. B., Pouvesle, J.-M., Rutkowski, R., Schuster, M., Bekeschus, S., Wende, K., Masur, K., Hasse, S., Gerling, T., Hori, M., Tanaka, H., Choi, E. H., Weltmann, K.-D., Metelmann, P. H., Von Hoff, D. D. and von Woedtke, T. (2018) Clinical experience with cold plasma in the treatment of locally advanced head and neck cancer. Clin. Plasma Med. 9, 6-13.
    CrossRef
  154. Mihai, C.-T., Mihaila, I., Pasare, M. A., Pintilie, R. M., Ciorpac, M. and Topala, I. (2022) Cold atmospheric plasma-activated media improve paclitaxel efficacy on breast cancer cells in a combined treatment model. Curr. Issues Mol. Biol. 44, 1995-2014.
    Pubmed KoreaMed CrossRef
  155. Min, T., Xie, X., Ren, K., Sun, T., Wang, H., Dang, C. and Zhang, H. (2022) Therapeutic effects of cold atmospheric plasma on solid tumor. Front. Med. (Lausanne) 9, 884887.
    Pubmed KoreaMed CrossRef
  156. Mirpour, S., Piroozmand, S., Soleimani, N., Jalali Faharani, N., Ghomi, H., Fotovat Eskandari, H., Sharifi, A. M., Mirpour, S., Eftekhari, M. and Nikkhah, M. (2016) Utilizing the micron sized non-thermal atmospheric pressure plasma inside the animal body for the tumor treatment application. Sci. Rep. 6, 29048.
    Pubmed KoreaMed CrossRef
  157. Mokhtari, H., Farahmand, L., Yaserian, K., Jalili, N. and Majidzadeh-A, K. (2019) The antiproliferative effects of cold atmospheric plasma-activated media on different cancer cell lines, the implication of ozone as a possible underlying mechanism. J. Cell. Physiol. 234, 6778-6782.
    Pubmed CrossRef
  158. Nagaya, M., Hara, H., Kamiya, T. and Adachi, T. (2019) Inhibition of NAMPT markedly enhances plasma-activated medium-induced cell death in human breast cancer MDA-MB-231 cells. Arch. Biochem. Biophys. 676, 108155.
    Pubmed CrossRef
  159. Nitsch, A., Strakeljahn, S., Jacoby, J. M., Sieb, K. F., Mustea, A., Bekeschus, S., Ekkernkamp, A., Stope, M. B. and Haralambiev, L. (2022) New approach against chondrosoma cells-cold plasma treatment inhibits cell motility and metabolism, and leads to apoptosis. Biomedicines 10, 688.
    Pubmed KoreaMed CrossRef
  160. Pan, M., Qin, C. and Han, X. (2021) Lipid metabolism and lipidomics applications in cancer research. Adv. Exp. Med. Biol. 1316, 1-24.
    Pubmed KoreaMed CrossRef
  161. Park, S.-B., Kim, B., Bae, H., Lee, H., Lee, S., Choi, E. H. and Kim, S. J. (2015) Differential epigenetic effects of atmospheric cold plasma on MCF-7 and MDA-MB-231 breast cancer cells. PLoS One 10, e0129931.
    Pubmed KoreaMed CrossRef
  162. Park, S., Kim, H., Ji, H. W., Kim, H. W., Yun, S. H., Choi, E. H. and Kim, S. J. (2019) Cold atmospheric plasma restores paclitaxel sensitivity to paclitaxel-resistant breast cancer cells by reversing expression of resistance-related genes. Cancers (Basel) 11, 2011.
    Pubmed KoreaMed CrossRef
  163. Partecke, L. I., Evert, K., Haugk, J., Doering, F., Normann, L., Diedrich, S., Weiss, F.-U., Evert, M., Huebner, N. O., Guenther, C., Heidecke, C. D., Kramer, A., Bussiahn, R., Weltmann, K.-D., Pati, O., Bender, C. and von Bernstorff, W. (2012) Tissue tolerable plasma (TTP) induces apoptosis in pancreatic cancer cells in vitro and in vivo. BMC Cancer 12, 473.
    Pubmed KoreaMed CrossRef
  164. Pasqual-Melo, G., Nascimento, T., Sanches, L. J., Blegniski, F. P., Bianchi, J. K., Sagwal, S. K., Berner, J., Schmidt, A., Emmert, S., Weltmann, K.-D., von Woedtke, T., Gandhirajan, R. K., Cecchini, A. L. and Bekeschus, S. (2020) Plasma treatment limits cutaneous squamous cell carcinoma development in vitro and in vivo. Cancers (Basel) 12, 1993.
    Pubmed KoreaMed CrossRef
  165. Pefani-Antimisiari, K., Athanasopoulos, D. K., Marazioti, A., Sklias, K., Rodi, M., de Lastic, A.-L., Mouzaki, A., Svarnas, P. and Antimisiaris, S. G. (2021) Synergistic effect of cold atmospheric pressure plasma and free or liposomal doxorubicin on melanoma cells. Sci. Rep. 11, 14788.
    Pubmed KoreaMed CrossRef
  166. Privat-Maldonado, A., Gorbanev, Y., Dewilde, S., Smits, E. and Bogaerts, A. (2018) Reduction of human glioblastoma spheroids using cold atmospheric plasma: the combined effect of short- and long-lived reactive species. Cancers (Basel) 10, 394.
    Pubmed KoreaMed CrossRef
  167. Privat-Maldonado, A., Verloy, R., Cardenas Delahoz, E., Lin, A., Vanlanduit, S., Smits, E. and Bogaerts, A. (2022) Cold atmospheric plasma does not affect stellate cells phenotype in pancreatic cancer tissue in ovo. Int. J. Mol. Sci. 23, 1954.
    Pubmed KoreaMed CrossRef
  168. Qi, M., Xu, D., Wang, S., Li, B., Peng, S., Li, Q., Zhang, H., Fan, R., Chen, H. and Kong, M. G. (2022a) In vivo metabolic analysis of the anticancer effects of plasma-activated saline in three tumor animal models. Biomedicines 10, 528.
    Pubmed KoreaMed CrossRef
  169. Qi, M., Zhao, X., Fan, R., Zhang, X., Peng, S., Xu, D. and Yang, Y. (2022b) Cold atmospheric plasma suppressed MM in vivo engraftment by increasing ROS and inhibiting the notch signaling pathway. Molecules 27, 5832.
    Pubmed KoreaMed CrossRef
  170. Rebl, H., Sawade, M., Hein, M., Bergemann, C., Wende, M., Lalk, M., Langer, P., Emmert, S. and Nebe, B. (2022) Synergistic effect of plasma-activated medium and novel indirubin derivatives on human skin cancer cells by activation of the AhR pathway. Sci. Rep. 12, 2528.
    Pubmed KoreaMed CrossRef
  171. Recek, N., Cheng, X., Keidar, M., Cvelbar, U., Vesel, A., Mozetic, M. and Sherman, J. (2015) Effect of cold plasma on glial cell morphology studied by atomic force microscopy. PLoS One 10, e0119111.
    Pubmed KoreaMed CrossRef
  172. Saadati, F., Mahdikia, H., Abbaszadeh, H.-A., Abdollahifar, M.-A., Khoramgah, M. S. and Shokri, B. (2018) Comparison of Direct and Indirect cold atmospheric-pressure plasma methods in the B16F10 melanoma cancer cells treatment. Sci. Rep. 8, 7689.
    Pubmed KoreaMed CrossRef
  173. Saadati, F., Moritz, J., Berner, J., Freund, E., Miebach, L., Helfrich, I., Stoffels, I., Emmert, S. and Bekeschus, S. (2021) Patient-derived human basal and cutaneous squamous cell carcinoma tissues display apoptosis and immunomodulation following gas plasma exposure with a certified argon jet. Int. J. Mol. Sci. 22, 11446.
    Pubmed KoreaMed CrossRef
  174. Sato, Y., Yamada, S., Takeda, S., Hattori, N., Nakamura, K., Tanaka, H., Mizuno, M., Hori, M. and Kodera, Y. (2018) Effect of plasma-activated lactated Ringer's solution on pancreatic cancer cells in vitro and in vivo. Ann. Surg.. Oncol. 25, 299-307.
    Pubmed CrossRef
  175. Schneider, C., Arndt, S., Zimmermann, J. L., Li, Y., Karrer, S. and Bosserhoff, A. K. (2018a) Cold atmospheric plasma treatment inhibits growth in colorectal cancer cells. Biol. Chem. 400, 111-122.
    Pubmed CrossRef
  176. Schneider, C., Gebhardt, L., Arndt, S., Karrer, S., Zimmermann, J. L., Fischer, M. J. M. and Bosserhoff, A.-K. (2018b) Cold atmospheric plasma causes a calcium influx in melanoma cells triggering CAP-induced senescence. Sci. Rep. 8, 10048.
    Pubmed KoreaMed CrossRef
  177. Schneider, C., Gebhardt, L., Arndt, S., Karrer, S., Zimmermann, J. L., Fischer, M. J. M. and Bosserhoff, A.-K. (2019) Acidification is an essential process of cold atmospheric plasma and promotes the anti-cancer effect on malignant melanoma cells. Cancers (Basel) 11, 671.
    Pubmed KoreaMed CrossRef
  178. Schuster, M., Seebauer, C., Rutkowski, R., Hauschild, A., Podmelle, F., Metelmann, C., Metelmann, B., von Woedtke, T., Hasse, S., Weltmann, K.-D. and Metelmann, H.-R. (2016) Visible tumor surface response to physical plasma and apoptotic cell kill in head and neck cancer. J. Craniomaxillofac. Surg. 44, 1445-1452.
    Pubmed CrossRef
  179. Senga, S. S. and Grose, R. P. (2021) Hallmarks of cancer-the new testament. Open Biol. 11, 200358.
    Pubmed KoreaMed CrossRef
  180. Sersenova, D., Machala, Z., Repiska, V. and Gbelcova, H. (2021) Selective apoptotic effect of plasma activated liquids on human cancer cell lines. Molecules 26, 4254.
    Pubmed KoreaMed CrossRef
  181. Shaw, P., Kumar, N., Hammerschmid, D., Privat-Maldonado, A., Dewilde, S. and Bogaerts, A. (2019) Synergistic effects of melittin and plasma treatment: a promising approach for cancer therapy. Cancers (Basel) 11, 1109.
    Pubmed KoreaMed CrossRef
  182. Silva-Teixeira, R., Laranjo, M., Lopes, B., Almeida-Ferreira, C., Gonçalves, A. C., Rodrigues, T., Matafome, P., Sarmento-Ribeiro, A. B., Caramelo, F. and Botelho, M. F. (2021) Plasma activated media and direct exposition can selectively ablate retinoblastoma cells. Free Radic. Biol. Med. 171, 302-313.
    Pubmed CrossRef
  183. Siu, A., Volotskova, O., Cheng, X., Khalsa, S. S., Bian, K., Murad, F., Keidar, M. and Sherman, J. H. (2015) Differential effects of cold atmospheric plasma in the treatment of malignant glioma. PLoS One 10, e0126313.
    Pubmed KoreaMed CrossRef
  184. Soni, V., Adhikari, M., Simonyan, H., Lin, L., Sherman, J. H., Young, C. N. and Keidar, M. (2021) In vitro and in vivo enhancement of temozolomide effect in human glioblastoma by non-invasive application of cold atmospheric plasma. Cancers (Basel) 13, 4485.
    Pubmed KoreaMed CrossRef
  185. Stoffels, E., Flikweert, A. J., Stoffels, W. W. and Kroesen, G. M. W. (2002) Plasma needle: a non-destructive atmospheric plasma source for fine surface treatment of (bio)materials. Plasma Sources Sci. Technol. 11, 383.
    CrossRef
  186. Stope, M. B., Benouahi, R., Sander, C., Haralambiev, L., Nitsch, A., Egger, E. and Mustea, A. (2020) Protherapeutic effects and inactivation of mammary carcinoma cells by a medical argon plasma device. Anticancer Res. 40, 6205-6212.
    Pubmed CrossRef
  187. Suzuki-Karasaki, M., Ando, T., Ochiai, Y., Kawahara, K., Suzuki-Karasaki, M., Nakayama, H. and Suzuki-Karasaki, Y. (2022) Air plasma-activated medium evokes a death-associated perinuclear mitochondrial clustering. Int. J. Mol. Sci. 23, 1124.
    Pubmed KoreaMed CrossRef
  188. Tahmasebi, G., Eslami, E., Naserzadeh, P., Seydi, E. and Pourahmad, J. (2020) Role of mitochondria and lysosomes in the selective cytotoxicity of cold atmospheric plasma on retinoblastoma cells. Iran. J. Pharm. Res. 19, 203-215.
  189. Tanaka, H., Mizuno, M., Katsumata, Y., Ishikawa, K., Kondo, H., Hashizume, H., Okazaki, Y., Toyokuni, S., Nakamura, K., Yoshikawa, N., Kajiyama, H., Kikkawa, F. and Hori, M. (2019) Oxidative stress-dependent and -independent death of glioblastoma cells induced by non-thermal plasma-exposed solutions. Sci. Rep. 9, 13657.
    Pubmed KoreaMed CrossRef
  190. Tavares-da-Silva, E., Pereira, E., Pires, A. S., Neves, A. R., Braz-Guilherme, C., Marques, I. A., Abrantes, A. M., Gonçalves, A. C., Caramelo, F., Silva-Teixeira, R., Mendes, F., Figueiredo, A. and Botelho, M. F. (2021) Cold atmospheric plasma, a novel approach against bladder cancer, with higher sensitivity for the high-grade cell line. Biology (Basel) 10, 41.
    Pubmed KoreaMed CrossRef
  191. Terefinko, D., Dzimitrowicz, A., Bielawska-Pohl, A., Klimczak, A., Pohl, P. and Jamroz, P. (2021) The influence of cold atmospheric pressure plasma-treated media on the cell viability, motility, and induction of apoptosis in human non-metastatic (MCF7) and metastatic (MDA-MB-231) breast cancer cell lines. Int. J. Mol. Sci. 22, 3855.
    Pubmed KoreaMed CrossRef
  192. Thiem, A., Has, C., Diem, A., Klausegger, A., Hamm, H. and Emmert, S. (2022) Wound therapy with cold atmospheric plasma in severe recessive dystrophic epidermolysis bullosa: a pilot study. Hautarzt 73, 384-390.
    Pubmed CrossRef
  193. Thiyagarajan, M., Sarani, A. and Gonzales, X. F. (2013) Characterization of an atmospheric pressure plasma jet and its applications for disinfection and cancer treatment. Stud. Health Technol. Inform. 184, 443-449.
    Pubmed
  194. Tian, Y., Zhang, Z., Zhang, Z. and Dai, X. (2022) Hsa_circRNA_0040462: a sensor of cells' response to CAP treatment with double-edged roles on breast cancer malignancy. Int. J. Med. Sci. 19, 640-650.
    Pubmed KoreaMed CrossRef
  195. Tornin, J., Labay, C., Tampieri, F., Ginebra, M. P. and Canal, C. (2021) Evaluation of the effects of cold atmospheric plasma and plasma-treated liquids in cancer cell cultures. Nat. Protoc. 16, 2826-2850.
    Pubmed CrossRef
  196. Tornin, J., Mateu-Sanz, M., Rodríguez, A., Labay, C., Rodríguez, R. and Canal, C. (2019) Pyruvate plays a main role in the antitumoral selectivity of cold atmospheric plasma in osteosarcoma. Sci. Rep. 9, 10681.
    Pubmed KoreaMed CrossRef
  197. Tornín, J., Villasante, A., Solé-Martí, X., Ginebra, M.-P. and Canal, C. (2021) Osteosarcoma tissue-engineered model challenges oxidative stress therapy revealing promoted cancer stem cell properties. Free Radic. Biol. Med. 164, 107-118.
    Pubmed KoreaMed CrossRef
  198. Trzeciak, E. R., Zimmer, N., Gehringer, I., Stein, L., Graefen, B., Schupp, J., Stephan, A., Rietz, S., Prantner, M. and Tuettenberg, A. (2022) Oxidative stress differentially influences the survival and metabolism of cells in the melanoma microenvironment. Cells 11, 930.
    Pubmed KoreaMed CrossRef
  199. Turrini, E., Laurita, R., Stancampiano, A., Catanzaro, E., Calcabrini, C., Maffei, F., Gherardi, M., Colombo, V. and Fimognari, C. (2017) Cold atmospheric plasma induces apoptosis and oxidative stress pathway regulation in T-lymphoblastoid leukemia cells. Oxid. Med. Cell. Longev. 2017, 4271065.
    Pubmed KoreaMed CrossRef
  200. Van, der Paal, J., Verheyen, C., Neyts, E. C. and Bogaerts, A. (2017) Hampering effect of cholesterol on the permeation of reactive oxygen species through phospholipids bilayer: possible explanation for plasma cancer selectivity. Sci. Rep. 7, 39526.
    Pubmed KoreaMed CrossRef
  201. Van Loenhout, J., Flieswasser, T., Freire Boullosa, L., De Waele, J., Van Audenaerde, J., Marcq, E., Jacobs, J., Lin, A., Lion, E., Dewitte, H., Peeters, M., Dewilde, S., Lardon, F., Bogaerts, A., Deben, C. and Smits, E. (2019) Cold atmospheric plasma-treated pbs eliminates immunosuppressive pancreatic stellate cells and induces immunogenic cell death of pancreatic cancer cells. Cancers (Basel) 11, 1597.
    Pubmed KoreaMed CrossRef
  202. Van Loenhout, J., Freire Boullosa, L., Quatannens, D., De Waele, J., Merlin, C., Lambrechts, H., Lau, H. W., Hermans, C., Lin, A., Lardon, F., Peeters, M., Bogaerts, A., Smits, E. and Deben, C. (2021) Auranofin and cold atmospheric plasma synergize to trigger distinct cell death mechanisms and immunogenic responses in glioblastoma. Cells 10, 2936.
    Pubmed KoreaMed CrossRef
  203. Vaquero, J., Judée, F., Vallette, M., Decauchy, H., Arbelaiz, A., Aoudjehane, L., Scatton, O., Gonzalez-Sanchez, E., Merabtene, F., Augustin, J., Housset, C., Dufour, T. and Fouassier, L. (2020) Cold-atmospheric plasma induces tumor cell death in preclinical in vivo and in vitro models of human cholangiocarcinoma. Cancers (Basel) 12, 1280.
    Pubmed KoreaMed CrossRef
  204. Vejdani Noghreiyan, A., Imanparast, A., Shayesteh Ara, E., Soudmand, S., Vejdani Noghreiyan, V. and Sazgarnia, A. (2020) In-vitro investigation of cold atmospheric plasma induced photodynamic effect by Indocyanine green and Protoporphyrin IX. Photodiagnosis Photodyn. Ther. 31, 101822.
    Pubmed CrossRef
  205. Vijayarangan, V., Delalande, A., Dozias, S., Pouvesle, J. M., Robert, E. and Pichon, C. (2020) New insights on molecular internalization and drug delivery following plasma jet exposures. Int. J. Pharm. 589, 119874.
    Pubmed CrossRef
  206. Von Woedtke, T., Schmidt, A., Bekeschus, S., Wende, K. and Weltmann, K.-D. (2019) Plasma medicine: a field of applied redox biology. In Vivo 33, 1011-1026.
    Pubmed KoreaMed CrossRef
  207. Walk, R. M., Snyder, J. A., Srinivasan, P., Kirsch, J., Diaz, S. O., Blanco, F. C., Shashurin, A., Keidar, M. and Sandler, A. D. (2013) Cold atmospheric plasma for the ablative treatment of neuroblastoma. J. Pediatr.. Surg. 48, 67-73.
    Pubmed CrossRef
  208. Wandke, D., Busse, B. and Helmke, A. (2022) Textbook of Good Clincial Practice in Cold Plasma Therapy. Switzerland Springer Nature.
  209. Wang, D., Zhang, J., Cai, L. and Dai, X. (2022a) Cold atmospheric plasma conveys selectivity against hepatocellular carcinoma cells via triggering EGFR(Tyr1068)-mediated autophagy. Front. Oncol. 12, 895106.
    Pubmed KoreaMed CrossRef
  210. Wang, L., Yang, X., Yang, C., Gao, J., Zhao, Y., Cheng, C., Zhao, G. and Liu, S. (2019) The inhibition effect of cold atmospheric plasma-activated media in cutaneous squamous carcinoma cells. Future Oncol. 15, 495-505.
    Pubmed CrossRef
  211. Wang, M., Holmes, B., Cheng, X., Zhu, W., Keidar, M. and Zhang, L. G. (2013) Cold atmospheric plasma for selectively ablating metastatic breast cancer cells. PLoS One 8, e73741.
    Pubmed KoreaMed CrossRef
  212. Wang, P., Zhou, R., Thomas, P., Zhao, L., Zhou, R., Mandal, S., Jolly, M. K., Richard, D. J., Rehm, B. H. A., Ostrikov, K. K., Dai, X., Williams, E. D. and Thompson, E. W. (2021) Epithelial-to-mesenchymal transition enhances cancer cell sensitivity to cytotoxic effects of cold atmospheric plasmas in breast and bladder cancer systems. Cancers (Basel) 13, 2889.
    Pubmed KoreaMed CrossRef
  213. Wang, P., Zhou, R., Zhou, R., Recek, N., Prasad, K., Speight, R., Richard, D., Cullen, P. J., Thompson, E. W., Ostrikov, K. K. and Bazaka, K. (2020) Chemo-radiative stress of plasma as a modulator of charge-dependent nanodiamond cytotoxicity. ACS Appl. Bio. Mater. 3, 7202-7210.
    Pubmed CrossRef
  214. Wang, Y., Mang, X., Li, X., Cai, Z. and Tan, F. (2022b) Cold atmospheric plasma induces apoptosis in human colon and lung cancer cells through modulating mitochondrial pathway. Front. Cell Dev. Biol. 10, 915785.
    Pubmed KoreaMed CrossRef
  215. Wang, Y., Zhang, L. and Zhou, F. (2022c) Cuproptosis: a new form of programmed cell death. Cell Mol. Immunol. 19, 867-868.
    Pubmed KoreaMed CrossRef
  216. Weiss, M., Gümbel, D., Gelbrich, N., Brandenburg, L.-O., Mandelkow, R., Zimmermann, U., Ziegler, P., Burchardt, M. and Stope, M. B. (2015a) Inhibition of cell growth of the prostate cancer cell model LNCaP by cold atmospheric plasma. In Vivo 29, 611-616.
    CrossRef
  217. Weiss, M., Gümbel, D., Hanschmann, E.-M., Mandelkow, R., Gelbrich, N., Zimmermann, U., Walther, R., Ekkernkamp, A., Sckell, A., Kramer, A., Burchardt, M., Lillig, C. H. and Stope, M. B. (2015b) Cold atmospheric plasma treatment induces anti-proliferative effects in prostate cancer cells by redox and apoptotic signaling pathways. PLoS One 10, e0130350.
    Pubmed KoreaMed CrossRef
  218. Welz, C., Emmert, S., Canis, M., Becker, S., Baumeister, P., Shimizu, T., Morfill, G. E., Harréus, U. and Zimmermann, J. L. (2015) Cold atmospheric
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles
  • CrossRef (0)

Funding Information

Services
Social Network Service

e-submission

Archives