You are on page 1of 10

Clinical Plasma Medicine 4 (2016) 19–28

Contents lists available at ScienceDirect

Clinical Plasma Medicine


journal homepage: www.elsevier.com/locate/cpme

Invited paper

The plasma jet kINPen – A powerful tool for wound healing


Sander Bekeschus n, Anke Schmidt, Klaus-Dieter Weltmann, Thomas von Woedtke
Leibniz-Institute for Plasma Science and Technology (INP Greifswald), ZIK plasmatis, Felix-Hausdorff Str. 2, 17489 Greifswald, Germany

art ic l e i nf o a b s t r a c t

Article history: The development of cold atmospheric pressure plasma sources was the starting point for the innovative
Received 8 August 2015 field of plasma medicine many years ago. Today, a large body of information is available on the bio-
Received in revised form medical and clinical applications of plasma. Among the latter, wound healing is of special interest as
15 January 2016
there have been promising studies demonstrating a benefit of plasma in the treatment of chronic
Accepted 19 January 2016
wounds. Wound healing is tightly regulated by redox mechanisms. This creates an exciting opportunity
as we and others have identified reactive oxygen and nitrogen species to be the central components
Keywords: mediating biological effects of cold plasma. A directed delivery of the plasma-generated species to cells
Atmospheric pressure argon plasma jet and tissues may therefore commence new therapeutic options not only in the healing of pathological
Chronic wounds
wounds but also related to other redox-based diseases. The literature covers numerous findings on the
kINPen MED
biological effects of cold physical plasma sources. However, plasma sources strongly differ from each
Plasma medicine
Reactive oxygen and nitrogen species other making it challenging to summarize or even compare results from different types of sources from a
Redox modulation physical and biological point of view. The aim of this comprehensive review is to provide a unique
compendium of studies assessing the pre-clinical and clinical relevance as well as potential health risks
related to the exposure to the atmospheric pressure argon plasma jet kINPen. This well-investigated and
promising plasma source demonstrates a strong potential to be of clinical significance in the near future.
Also, the collection of studies investigating its biological effects may serve as a role model for similar
plasma sources and applications in the field of plasma medicine.
& 2016 Elsevier GmbH. All rights reserved.

1. Introduction 20]. Recently, also an imperative role for plasmas in cancer treat-
ment has been implied [21–30]. Another highly promising area of
1.1. Plasma medicine application is treatment of chronic wounds to improve their
healing which is the focus of the present work [31–33]. However,
The innovative field of plasma medicine is emerging as a hot plasma sources strongly differ from each other making it chal-
topic, not only in the community of physics. For over 20 years now lenging to summarize or even compare results from different
[1], cold plasma is being investigated for biomedical applications types of sources from a biological or physical point of view. This
using appropriate laboratory testing. Yet, significant progress on compendium will focus on a particularly promising argon plasma
its molecular mechanisms has only been made in the past decade jet (kINPen; Leibniz Institute for Plasma Science and Technology –
[2]. The role of oxidants and antioxidants in cells and tissues re- INP Greifswald and neoplas tools GmbH Greifswald, Germany) and
lating to the effects of plasma has increasingly been pinpointed in the pre-clinical and clinical studies obtained with this device.
recent redox biological work [3–5]. Also, an ever increasing body Potential health risks in humans as a consequence of the plasma
of information is available on controlling and tuning the plasma application using the kINPen will also be discussed. Numerous
studies were concerned with this risk assessment as it is pivotal
and its reactive species output in need to modify physical and
for a general acceptance of cold physical plasmas among physi-
biology targets [6–8]. Today, cold plasmas are present in a variety
cians, clinicians, and patients alike.
of applications. This includes the decontamination of heat-sensi-
tive materials, such as, surgical equipment, without introducing
1.1.1. Cold physical plasma
alterations in their quality [9–11]. Food [12–14] and its packaging
Plasma is generated by energizing gas up to a critical point at
[15–17] has been decolonized using cold plasma as well. Plasma
which electrons dissociate from atoms. The resulting ionized gas
can contribute to surface functionalization in implantology [18– contains charged particles, while the overall charge remains
electrically neutral. In biomedical applications, plasma sources
n
Corresponding author. that operate at atmospheric pressure and transfer a minimal
E-mail address: sander.bekeschus@inp-greifswald.de (S. Bekeschus). thermal output are desirable in order to be tolerated by cells and

http://dx.doi.org/10.1016/j.cpme.2016.01.001
2212-8166/& 2016 Elsevier GmbH. All rights reserved.
20 S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28

tissues during exposure. In plasma jets, such as the kINPen, the integrated mass flow controller, ii) a housing optimized for the
plasma is usually generated by applying a high-frequency alter- clinical use, and iii) a pulsed plasma generation (2.5 kHz, plasma
nating voltage to a gas. The electron flux exhibits a high velocity on to off ratio ¼1:1). In the kINPen MED, the latter further reduces
and is hot. These fast electrons then excite or ionize atoms and/or the amount of energy being transmitted, i.e., the temperature at
molecules of a feed gas, e.g., argon. Argon ions on the other hand the tip of the effluent and the UV output. Despite the lower energy
are heavier than electrons and hence slower in the electric field, out, the biological effects of both jets were found to similar in
preventing their acceleration. Also, fast electrons inefficiently HaCaT keratinocytes in vitro [42]. Hydrogen peroxide seems to
transfer their energy (heat) to the heavy particles which conse- dominate the liquid chemistry of a number of plasma sources in-
quently remain cold. As the heavy particle temperature of a gas cluding the kINPen [43–50]. It was found to be deposited in similar
determines its overall temperature, the plasma contains highly concentrations in liquids by both the kINPen MED and the kINPen
energized particles without displaying the significant temperature 09 (unpublished observation), possibly explaining their compar-
increase that would usually come with them (non-equilibrium able effects in vitro. Yet, these preliminary results contradict ex-
plasma). Additional cooling of plasma can be achieved using pectations with regard to the different operation modes (con-
pulsed excitation patterns or high feed gas fluxes, e.g., of argon. tinuous vs. pulsed) and further in depth analysis is needed to ex-
The feed gas flux also provides a channel of easy-to-ignite gas at amine the similarities and differences regarding the reactive spe-
the outside of the jet nozzle [34]. The plasma being generated in cies output between the two related kINPen devices. Central
this effluent region interacts with ambient air to create non-ra- properties of the kINPen 09 and the kINPen MED are summarized
dical (e.g., hydrogen peroxide, ozone) or radical species (e.g., hy- in Table 1. The kINPen MED (Leibniz Institute for Plasma Science
droxyl radical, nitrogen monoxide) [35] which then oxidize and Technology – INP Greifswald and neoplas tools GmbH
biomolecules. Greifswald, Germany) was the first cold atmospheric pressure
plasma jet worldwide to be accredited as a medical device (class
1.1.2. The kINPen argon plasma jet IIa) for the use in patients. This argon plasma jet is a reusable
The kINPen is a commercially available atmospheric pressure device with the purpose of generating a constant, non-thermal
argon plasma jet. Its concentric, light-weighted, and pen-like de- (room-temperature) plasma at atmospheric pressure. Its use is
sign was developed for biomedical applications and allows precise intended for the treatment of non-healing wounds and ulcers of
and arbitrary 3D movements [36]. Typical active agents being the human skin. The compromised tissue may be infected and the
generated include ions, electrons, or reactive oxygen and nitrogen plasma can be effectively applied regardless of any bacterial
species (ROS/RNS). Electric and magnetic fields, light (visible, in- multidrug resistances.
frared, UV), and neutral particles are also being generated. The
hand-held unit is operated using a power supply and a gas supply 1.2. Wound healing
unit. The electrical safety of the kINPen is certified and complies
with EU standards (certificate number 609.003.1). In its metal In life, tissue injury is inevitable. Microorganisms may invade
housing, a central, rod-like electrode is mounted and shielded by a compromised tissues and cause infection. Tissues display a high
dielectric quartz capillary connected to a grounded ring electrode. plasticity and the ability to seal a gap, while bacteria are being
The plasma is generated by applying a sinusoidal voltage removed by immune cells: to heal.
(2–6 kVpp) with a frequency of 1.0–1.1 MHz to the central electrode
(power: o3.5 W in the hand-held unit). The typical plasma ef- 1.2.1. Healing phases
fluent length is 9–12 mm and with 1 mm in diameter. Argon is The process of wound healing is characterized by four con-
used as feed gas, but small amounts of other molecular gases can tinuous, overlapping, and precisely programmed phases: hemos-
be admixed [37]. High spatial and temporal optical resolution of tasis, inflammation, proliferation, and remodeling [51]. Hemostasis
the plasma revealed that it propagates in a bullet-like manner is characterized by vascular constriction and platelet aggregation,
[38]. If applied as recommended, temperature or UV-radiation are degranulation, and fibrin formation (thrombus) [52]. In the sub-
not harmful to humans [36,39]. Recently, a gas curtain was in- sequent inflammatory phase, neutrophils infiltrate the wound site,
troduced, shielding the plasma from ambient air [40]. This allows followed by monocytes and lymphocytes [53]. During the pro-
for a more refined control of the environmental conditions sur- liferation phase, re-epithelialization occurs, while new blood ves-
rounding the plasma and influencing its chemistry [41]. The kIN- sels are being formed (angiogenesis) and collagen is being syn-
Pen MED (Fig. 1) is a clinic-oriented evolution of its predecessor, thesized for extracellular matrix formation [54]. Finally, collagen
the kINPen 09. Both units are identical in essential respects. The and the vasculature are being remodeled [55]. Many different cell
kINPen MED mainly differs from the kINPen 09 by having i) an types are involved in the healing process, such as, fibroblasts and

Fig. 1. The kINPen MED.


S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28 21

Table 1 through the normal stages of healing and therefore enter a state of
Physical properties of the kINPen 09 and the kINPen MED. pathologic inflammation” [74]. It is possible that different parts of a
wound may be stuck in different healing phases, having lost the ideal
Properties kINPen 09 kINPen MED
synchrony of events [75]. Consequently, healing is delayed and in-
Recommended feed gas Argon (5 7 1 l/min) argon (5 7 1 l/min) complete, resulting in poor anatomical and functional outcome.
and flux Chronic wound etiology is heterogeneous but most ulcers are caused
Frequency 1.1 MHz 1 MHz by ischemia, secondary to diabetes mellitus, venous stasis, and
Feed gas flow control Via external devices Yes (integrated)
Mode of operation Continuous Pulsed (2.5 kHz)
pressure. Additionally, wound healing can be compromised by in-
Duty cycle n. a. Plasma on/off ¼1:1 fection with microorganisms [76]. In healing wounds, inflammatory
Power o 8 W of the whole o8 W of the whole processes effectively eradicate microbial invasion whereas in dete-
system system riorated tissues (ischemia, hypoxia, devitalized tissue, and chronic
o 3.5 W of the handheld o3.5 W of the hand-
inflammation) this process may be impeded. Consequently, in-
device held device
Typical effluent length 10–12 mm 9–11 mm flammation, e.g., mediated by neutrophils, cannot be resolved. In
Recommended working 9–11 mm 8–10 mm normal healing, neutrophil presence is self-limited to 72–96 h after
distance wounding [77]; in non-healing wounds they are constantly present
Temperature at the re- 46–50 °C 35–38 °C
[78]. A continuous presence of neutrophils leads to an imbalance in
commended working
distance
proteinase and cytokine concentrations. Proteinases degrade tissue,
UV-irradiation at work- UVA: 20–35 mW/cm2 UVA: 5–15 mW/cm2 and this proteolytic environment disallows the formation of suffi-
ing distance (free jet) UVB: 20–15 mW/cm2 UVB: 5–15 mW/cm2 cient extracellular matrix, thus inhibiting cell migration and collagen
Phase stabilization dur- Manually Automatic: PLL-circuit deposition [79]. These conditions seem to be an indirect result of an
ing plasma generation
amplified cytokine secretion. Unfortunately, to date there is no gen-
erally accepted chronic wound animal model available [80], render-
ing animal experiments difficult to interpret. This applies, for ex-
keratinocytes [56]. For wound closure, these cells are essential for ample, to studies addressing the role of bacteria in improper wound
extracellular matrix deposition, and tissue remodeling [57]. Se- healing. While presence of bacteria is a well-recognized factor in
quential immigration of immune cells helps to eradicate infectious chronic wounds [81] their eradication is not necessarily associated
agents and to orchestrate the wound healing phases [58]. Espe- with improved healing responses [82]. Nonetheless, their removal is
cially macrophages pave the way for wound resolution by pha- a prerequisite for wound stabilization to allow for wound healing
gocytosing dead cells, coordinating the healing response via re- phase progression.
lease of cytokines and growth factors [59], and promoting the
proliferative phase via fibroblast and keratinocyte stimulation as
well as VEGF-mediated angiogenesis [60]. 2. Pre-clinical studies using the kINPen

1.2.2. Redox control in wounds 2.1. In vitro activity of plasma against bacteria and other pathogens
For a long time, the dogma has been that reactive species are
primarily toxic to cells. Yet, further studies revealed that at low Similar to numerous other plasma sources [83–85], the kINPen
concentrations these species play a pivotal role in cellular phy- has been shown to be effective against various bacteria in vitro
siology and signaling [61]. They are also thought to act as second including Escherichia coli, Pseudomonas aeruginosa, Klebsiella group
messengers [62]. During the first event in wound healing, he- (K. pneumoniae, K. oxytoca), Staphylococcus aureus, hemolysing
mostasis, platelet aggregation occurs, and these cells are capable Lancefield Streptococci (group A and B), Proteus group (P. mirabilis,
of generating reactive oxygen species (ROS) [63]. At the same time, P. vulgaris), Acinetobacter spp., Stenotrophomonas spp., Enterococcus
scavenging of ROS inhibits collagen-dependent platelet aggrega- faecalis, and Staphylococcus epidermidis [86–91]. Biofilms can also
tion [64]. Phagocytes utilize oxidants for antimicrobial defense be successfully removed with this argon plasma jet [5,92–94]. In a
[65]. Interestingly, stimulation with growth factors initiates ROS comprehensive study, the growth of 194 clinical isolates of chronic
production in many cell types [66]. Mutations in a ROS-generating wounds from 13 different and partly multidrug-resistant species
enzyme, the NADPH oxidase, causes the chronic granulomatous was tested. All organisms very successfully inactivated using the
disease which is an immune-deficient condition characterized by kINPen [95]. The plasma of the kINPen was also shown to be ef-
impaired wound healing [67]. Hydrogen peroxide (H2O2) induces fective against the EHEC pathogen [96]. To test the efficacy of the
chemotaxis in neutrophil granulocytes to the site of infection [68]. antiseptic effects of plasma on animal tissue, freshly enucleated
H2O2 is especially interesting as it has a fine-tuning regulatory role porcine eyes were used as a test model. Compared to two com-
in both inflammation as well as avoidance of harmful in- monly used antiseptics, the plasma treatment was significantly
flammatory responses [69]. ROS can induce epithelial cell pro- more effective against both S. aureus and P. aeruginosa [97]. Fungi
liferation and migration which are both important in wound re- and parasites were also successfully inactivated using the plasma
epithelialization [70]. To date, the highest levels of H2O2 in the of the kINPen. In vitro, this includes the clinical fungal strain in-
body were found in wound fluids [71] with physiological con- volved in dermatomycosis, namely Trichophyton interdigitale, Tri-
centrations ranging between 100 and 250 mM [72]. Interestingly, chophyton rubrum, Microsporum canis, and Candida albicans [98–
these concentrations were modulated during different healing 100]. Plasma treatment also kills wound-causing mites [101]. Al-
phases which again suggest a physiological role of oxidants and together, these data suggest the plasma of the kINPen to be useful
antioxidants in wound healing. Thus, ROS are central in wound in the treatment of infected and/or chronic wounds, especially if
healing, and this creates an exciting therapeutic window for cold multiresistant pathogens are governing the infection that are
physical plasma-generated reactive species in wound treatment. usually difficult to treat using conventional therapies.

1.2.3. Non-healing and chronic wounds 2.2. Redox stimulation of skin and immune cells using plasma
Non-healing wounds are a major health issue in the western
world, causing an estimated $3 billion per year in the US [73]. By The central function of skin cells in wounding is the wound
definition, non-healing or chronic wounds “failed to progress closure by cell division and migration [102]. Plasma treatment
22 S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28

improved the closure of an artificially created, infected wound in a in chronic wound healing as there are no valid animal models
2-D skin model [103]. Plasma exposure of keratinocytes in vitro available for this pathology [124–126]. However, wound healing can
also resulted in an enhanced production of growth factors im- be slowed for up to several weeks in a hairless mouse model [127–
portant for wound healing and neovascularization [104]. Also, 129]. Using this model, an animal study with 77 mice was conducted
plasma affected the keratinocyte transcriptome [105] and pro- to examine wound healing after plasma-treatment. Preliminary re-
teome [106] strongly via antioxidant pathways [107] in vitro. Both sults showed that the wound area on ears receiving plasma (3 s) at
in vitro [108] and ex vivo [109], plasma treatment enhanced the day 9 was about 30% smaller compared to untreated controls
proliferation rate of skin cells which is highly important for wound (manuscript in preparation). Moreover, healing of chronic wounds
closure. Therefore, the plasma of the kINPen alters the redox was markedly induced in domestic animals (4 dogs and 2 cats) [108].
balance in skin cells which positively affects their growth and The wound age was 2–60 months and some animals failed multiple
secretion of growth factors. During wound healing, immune cells treatments using conventional antiseptics or skin graft surgery.
orchestrate the wound healing phases and are decidedly required Within 3–24 weeks of regular plasma treatment (twice weekly),
for antibacterial defenses. After plasma treatment, the ability of complete wound healing was achieved. Altogether, plasma treatment
neutrophils to ingest and kill bacteria was not altered but overall with the kINPen supports the healing of slow-healing or truly chronic
antibacterial efficacy maybe increased due to extracellular trap wounds in animals.
formation [110] which effectively inhibits microbial growth [111].
This may decrease wound inflammation and would support 3.3. Enhanced wound healing in clinical observational studies
healing. Plasma was not toxic in neutrophils or monocytes [112]
but readily induced apoptosis in lymphocytes [113]. This was due The efficacy of the kINPen MED in the treatment of wounds has
to strong oxidation in the cell membrane and cytosol [114], cul- been tested in three observational studies with a total of 26 patients.
minating in an altered redox state in these cells [115]. The cyto- All studies demonstrated a wound healing and/or antiseptic effect of
toxic effects seen in lymphocytes are highly H2O2-dependent the plasma. In a clinical case-control study, the efficacy of the kINPen
which seems to be generated in the plasma gas and liquid phase for wound healing was compared to Octenisept which is one of the
[116, 117]. Elevated numbers of lymphocytes are present in pa- most effective and biocompatible antiseptics in the management of
thological wounds [118] and their removal via oxidants may be chronic wounds. Sixteen patients with chronic leg ulcers were en-
beneficial for healing. Nonetheless, lymphocytes are also im- rolled in this study. They received plasma three times a week and
portant to elicit immune responses, and these cells retained their over a total period of two weeks [130]. The number of bacterial co-
ability to proliferate after exposure to the plasma [119]. Plasma lonies and the size of the wound surface and were determined
stimulated monocytic cells, and induced inflammatory as well as thereafter. Treatment with either the kINPen MED or Octenisept
anti-inflammatory redox response at the same time [120]. These achieved a comparable reduction of bacteria. The wound surface re-
redox modulations may tip the balance of the low-grade in- duction was 56% in the plasma group and 19% in the Octenisept
flammation seen with chronic wounds towards the transition to group, suggesting a benefit of plasma in wound healing [131]. A
the proliferative phase required for proper wound healing. positive side effect of the plasma treatment was a slight reduction of
wound exudation. Exposure to plasma was tolerated very well. In
another case-report study, four sterile wounds were induced on both
3. Case reports and clinical results using the kINPen plasma forearms of five volunteers and utilizing a UltraPulse CO2 laser com-
monly used in esthetic surgery [132]. In a randomized, controlled
3.1. Antimicrobial efficacy experimental procedure, the wounds then either received plasma
treatment (single dose 10 s, three times 10 s, or single dose 30 s) or
One-hundred and five wound-resident bacteria and fungi (11 remained untreated for three consecutive days. Wound healing pro-
different species with some displaying multidrug resistances) were gress was photographed 10 days after wounding and staged in a
plasma-treated on contaminated finger tips of nine healthy volun- blinded manner by five independent physicians. They scored i) which
teers. Among the bacterial strains were Pseudomonas, Klebsiella, Sta- of the four wound sites presented the best early healing, and ii) how
phylococci, and Micrococci which are often difficult to treat in clinical should the esthetic outcome of this best site be scored on a numeric
settings. All types of bacteria and fungi were highly susceptible to scale (0–10 with 10 scoring the ideal result). Three times 10 s plasma
plasma treatment and regardless of them being part of the physio- treatment on three consecutive days gave the best results followed by
logical or pathological skin flora [121]. The tests were carried out in the single 30 s treatment. Ten seconds single treatment was similar to
accordance with the national guidelines for microbiological diag- untreated control. Importantly, healing was final and without com-
nostics, and the net reduction of bacterial burden was significant, plications as determined in a follow-up study 1 year after plasma
although not similar for all types of bacteria tested. The authors treatment [133]. In the third study, Vandersee and colleagues induced
suggest that the plasma of the kINPen may be an effective supple- four artificial and sterile wounds using negative pressure on the both
ment to antiseptics in the treatment of skin and wound-resident arms of five volunteers [134]. Initial wound size was 1.0–1.8 cm2. The
pathogens. The antimicrobial activity of the plasma on the skin was four wounds received either no treatment, treatment with the kIN-
confirmed in a study by Klebes and colleagues. Chronic wounds of 34 Pen MED for 60 s, treatment with Octenisept, or a combination
patients were treated with the plasma of the kINPen 09 or in com- therapy involving both regimens. Wounds receiving plasma displayed
bination with a wound antiseptic. The combination therapy showed an accelerated healing response while untreated wounds showed an
the best efficacy [122]. In another study involving six psoriatic pa- average healing response, respectively. Using Octenisept or the
tients, an antimicrobial efficacy of plasma was found as well but re- combination therapy, the average grade of wound healing was above
mission of the disease was not observed [123]. Thus, the in vivo ap- untreated control but below plasma treatment alone. Using confocal
plication of the plasma jet kINPen is effective against bacteria residing laser scanning microscopy, it was demonstrated that plasma-treated
on the skin and in wounds. wounds displayed an earlier onset of the proliferative wound healing
phase, possibly explaining the accelerated healing response observed.
3.2. Enhanced wound healing in animal studies This also implies a shortened inflammatory phase, suggestion a
plasma-induced redox modulation of immune cells that regulate in-
The efficacy of new therapeutic concepts is usually evaluated in flammation to be central. It can be concluded that plasma treatment
animals first. However, it is challenging to investigate improvement with the kINPen MED is beneficial for healing of chronic wounds.
S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28 23

Accelerated healing was also seen with sterile wounds, proposing a to be expected at constant (8 h) concentrations of 0.055 ppm in air
promotion of wound healing independent of the antimicrobial effects [145]. Ozone concentrations in hot, European summers are
previously seen with the plasma. Vice versa, the efficacy of plasma sometimes 0.3 ppm locally [146] but on average between 0.03 and
may be enhanced in infected wounds due to its antiseptic properties. 0.05 ppm. Short-term concentrations of 1.5 ppm are considered to
Therefore, treatment with the kINPen MED is an innovative approach be an acute health risk. The kINPen 09 expels maximum ozone
to improve healing of wounds. Yet, and similar important to its effi- concentrations of 0.10–0.13 ppm and this only in the immediate
cacy, the plasma treatment needs to be safe in order to achieve a vicinity of the plasma source (distance o10 cm) [141]. At larger
widespread clinical application in future. distances ( 430 cm) the ozone concentration is below 0.10 ppm
[39]. Because of the brief exposure during the rather short-term
plasma treatment, acute health risks via ozone formation are not
4. Risk estimation of the plasma of the kINPen to be expected. The odor threshold for ozone is 0.02 ppm and may
be exceeded during plasma treatment, possibly causing minor
4.1. Physicochemical parameters unease of odor. Formation of potentially toxic nitrogen dioxide was
not detected in the plasma effluent of the kINPen [36].
4.1.1. Temperature
An autoclavable spacer is available for the kINPen MED to en- 4.2. Biological parameters
sure a uniform plasma treatment. It facilitates a fixed distance (7–
8 mm) from the jet nozzle to the surface in question. For the 4.2.1. Mutagenicity
kINPen, an efficient oxidant delivery was shown for distances of up Cold plasma expels various reactive components which can
to 12 mm [135]. Within this distance, a thermal energy dissipation oxidize biomolecules and are therefore potentially mutagenic. A
of the plasma is still measureable. To avoid thermal denaturation common OECD test systems for genetic toxicology is the HPRT
of proteins in tissues, the temperature output of the jet needs to be gene mutation assay that utilizes the Chinese hamster cell line V79
acceptable. Using a thermal camera, the average temperature at [147]. In this test, viable mutants are detected by the loss of ac-
the clinically relevant distance of 7–8 mm was found to be tivity of the HRPT enzyme which generates cytotoxic nucleotides
39 71 °C [132]. On the skin of volunteers and by applying an ap- that otherwise kill the non-mutants. Neither exposure to plasma-
propriate velocity of the plasma jet over the skin (8 mm treated medium nor direct plasma treatment (30 s) induced an
per second), no temperature increase was observed (manuscript in increase of mutants [148]. Micronucleus formation is assessed in
preparation). The lack of thermal damage was verified histologi- another OECD test system for genetic toxicology [149]. Plasma
cally in plasma-treated porcine skin [136]. treatment did not induce the formation of micronuclei [148] in-
dicative of acentric DNA fragments that are common in mutations
4.1.2. UV radiation [150]. Also, non-mutated cells do not form colonies in soft agar
High doses of UV radiation can induce DNA double-strand breaks while malignant cells have often lost the need of cell–cell contact
and thereby pose a carcinogenic hazard. The International Commis- for proliferation. During 30 days after plasma treatment, HaCaT
sion on Non-Ionizing Radiation Protection (ICNIRP) [137] recommends keratinocytes did not form colonies in soft-agar which further
a maximum effective (weighted) exposure of 30 J/m2 per day (8 h; substantiates a lack of mutagenicity of the plasma of the kINPen
λ ¼ 180–400 nm) [138]. The minimum exposure needed to elicit skin [148]. In ex vivo treated human skin, the plasma of the kINPen
erythema (sun burn) in the most sensitive human skin type is MED did not induce double strand breaks which can be indicative
200 J/m2 [139]. By means of optical emission spectroscopy, it was of mutagenic events [151]. In a long-term clinical investigation,
found that the kINPen MED generates some UV emission in the UV-A artificial skin wounds in volunteers were plasma-treated and
and UV-B range, and very low emission in the UV-C range. In liquids, wound healing side effects were graded one year after treatment.
the latter was shown to be very quickly absorbed (100% absorption at In none of the five subjects a sustained tissue degeneration or
about 220 nm of liquid depth for cell culture medium treated with cancerous lesion was observed [133]. Preliminary results of mice
the kINPen 09) [140]. For plasma treatment (60 s of 0.5 cm2 at a that have received plasma treatment of wounds did not provide
distance of 7–8 mm), the weighted effective irradiance of a “free” any indication of a mutagenic action of plasma (manuscript in
kINPen MED plasma (not touching a surface) was measured and preparation). Although further research in animal models is nee-
calculated to be 1.05 J/m2 [141]. If the plasma jet is not being moved ded to verify or exclude a mutagenic role of cold plasma, such a
during the 60 s treatment time (single-spot exposure) the UV ex- health risk cannot be concluded from scientific studies to date.
posure is locally 8.8 J/m2. Both figures are well below the re-
commended limits [142]. Yet, it is known that the properties of jet 4.2.2. Penetration depth in the skin
plasmas may change dramatically if the effluent comes in contact The plasma treatment of the skin or wounds aims at setting
with a surface [143], very likely increasing electron densities and topical effects on the surface of the tissue. Subcutaneous tissues,
therefore also the reactive species and UV as well as VUV output. As such as, muscles or fat, should not be affected by the treatment.
such, further investigations are needed to better understand poten- Therefore, several studies determined the penetration depth of
tial risks of UV exposure of the kINPen plasma if in contact to a plasma in the skin. Plasma generates reactive species that have
substrate, such as skin. oxidizing properties in, e.g., β-carotenoids in intact skin, which can
be assessed using Raman microspectroscopy [152]. Plasma treat-
4.1.3. Generation of ozone ment of intact skin of six subjects showed oxidation to be present
During the plasma treatment with the kINPen, ozone is being only up to a depth of 10 mm [153]. Similar results were obtained in
formed [144]. Ozone is part of environmental technologies for air other work [154]. In both studies and compared to untreated skin,
and water disinfection, and is an unavoidable part of many other no significant differences were observed in deeper layers of the
technologies, such as, laser printers in offices. The previously skin. This implies that the oxidative challenge provided by the
common MAK (maximum working concentration value) for ozone kINPen plasma in tissues is only superficial.
was 0.1 ppm in air, and is defined as a maximum and permanent
concentration during a work day (8 h). Today, there are no binding 4.2.3. Subjective sensations
limits of its concentration in ambient air as ozone has been clas- Subjective sensations of the plasma treatment may not per se
sified as potentially carcinogenic. Nevertheless, no health risks are constitute a potential health risks but decrease the acceptance of
24 S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28

plasma in patients. After a single-spot plasma treatment of the mice studies underlined the potential of different cold plasma
fingertips of nine volunteers, these were asked to indicate pain, sources for accelerated wound healing [179–182]. Also, safety
heat, as well as other sensations (paresthesia) of the skin on a scale considerations were made for a number of sources using animal
from 0 (no sensation) to 10 (extreme sensation) [121]. The fol- skin [183–185]. While first efforts have been made to rule out
lowing results were obtained for a treatment time of 240 s using mutagenic effects using in vitro models [186], long-term animal
the kINPen: 0.5 (pain); 1.3 (heat); 1.5 (paresthesia). No volunteer studies supporting this notion have not been published yet for any
wished to discontinue the study due to any discomfort, such as, plasma source.
pain or heat. A similar study was conducted with artificially cre-
ated wounds (suction blisters) in five volunteers [134]. On a scale
of 0 (no irritation) to 5 (intolerably strong irritant effect) for sen- 5.2. Clinical results
sory irritations, the volunteers reported a 1 in average. Further
validating the results of ozone measurements with the kINPen, no Many plasma source have been developed for biomedical ap-
volunteer or physician reported an unpleasant smell of any ozone plications but only four (including the kINPen MED) received ac-
created during the treatment. It was concluded that the plasma creditation as a medical device for wound healing so far. Among
treatment is tolerated well subjectively. these is the recently (August 2015) marketed SteriPlas (AdTec Ltd.,
Japan) which was proven to be effective in reducing bacterial
4.2.4. Cytotoxicity and histocompatibility burden in wounds [187,188] and supported their healing [189,190]
Plasma treatment should neither irritate the skin nor cause as well as reduced local pain [191]. Another accredited source is
large-scale damage or toxic effects. For ethical reasons, such stu- the PlasmaDerm (Cinogy GmbH, Germany) that has been shown to
dies cannot be carried out in humans but animal tissues serve as a be safe and beneficial in the healing of chronic leg ulcers in a
good model to investigate the plasma's irritation potential. As a randomized and controlled clinical trial [192]. Also PlasmaOne
replacement method for the Draize test in the rabbit eye, the HET- (Medical Systems GmbH, Germany) is a plasma source accredited
CAM test (Hen's Egg Test on the chorioallantoic membrane) is as a medical device. There is a vast number of plasma therapy case
commonly used to examine the biocompatibility of chemicals. reports available but its efficacy in supporting wound healing or
Clinically-relevant exposure times to non-pulsed (kINPen 09) conferring antimicrobial activity has not been scientifically pub-
plasma caused an average hemorrhagic or intravasale thrombosis lished so far. Nonetheless, for each accredited device it has been a
on the CAM. By contrast, plasma treatment using the pulsed mode long way from research to market. Yet, and considering the stirring
(similar to the kINPen MED) was superior and resulted in only potential of cold plasma for the healing of chronic wounds in
slight irritation [155]. Strikingly, the irritation was further reduced millions of patients worldwide, it is worth any effort. There is
by a parallel application of hydrocortisone [156]. This underlines much reason to be excited about future plasma research and
the role of plasma-mediated redox modulation in immune cells, medical sources participating in tackling human skin pathologies.
and suggests plasma to be particularly effective in multimodal
therapies. Importantly, the irritant effect was completely re-
versible with clinically relevant plasma treatment times (60 s).
Next to the irritation potential of plasma, histological analysis was 6. Conclusion
carried in different studies and after plasma treatment. The plasma
of the kINPen showed identical histocompatibility compared to The atmospheric pressure argon plasma jet kINPen is among
two commonly used skin antiseptics in an experimental model of the most promising sources in the field of plasma medicine. Its
porcine eyes [97]. Histological analysis was also carried out in electrical safety is verified (CE-mark), it has received certification
surgically removed human skin which was exposed to the plasma as a medical device class IIa, and it has been proven to be anti-
of the kINPen MED for 60 s. Plasma treatment did not increase the septically effective. Prominently, it supports the healing of
number of apoptotic cells, did not induced epidermal lesions, and wounds. According to the available literature, its application does
did not elevate extracellular concentrations of cytochrome c which not pose any health risks in humans with regard to UV exposure,
is an marker for physical cell damage [109]. A lack of histological thermal damage, tissue toxicity, or mutagenicity. The biological
damage was also seen after 10 min of single-spot plasma treat- and medical investigations carried out with this device may serve
ment on human skin ex vivo [157]. In another study, no significant as role model for other plasma sources in the field of plasma
effect of the plasma on the water balance (moisture) of the skin medicine, and will stimulate curiosity and interest in the plasma
was found [158]. In sum, the cold plasma treatment with the technology altogether.
kINPen is histocompatible and not damaging to the skin per se.

5. Other sources for plasma medicine Conflict of interest

5.1. Preclinical studies Klaus-Dieter Weltmann is a minority shareholder and the INP
Greifswald is a majority shareholder of neoplas tools (Greifswald,
A vast body of information is available about preclinical studies Germany). All other authors declare no conflicts of interest.
using various types of cold plasma sources with similar anti-
microbial efficacy compared to the kINPen. These are effective
against microorganisms in the planktonic state [159–161], on agar Acknowledgments
[162–164], in biofilms [165–167], and drug-resistance pathogens
[168–170]. A hallmark of cold physical plasma is its proposed se- This work was funded by the German Federal Ministry of
lective efficacy against unwanted microorganisms while leaving Education and Research (Grant number 03Z2DN11) and by the
eukaryotic cells intact. Additionally, it has been observed that Ministry of Education, Science and Culture of the State of Meck-
wound-relevant processes are supported, including activation and/ lenburg-Western Pomerania and the European Union, European
or enhanced proliferation of fibroblasts [171–174], endothelial cells Social Fund under (Grant numbers AU 11 038 and ESF/IV-BM-B35-
[175], and keratinocytes [176–178]. Importantly, several in vivo in 0010/13).
S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28 25

References proliferative activity against chronic chemo-resistant ovarian cancer cells in


vitro and in vivo, PLoS One 8 (2013) e81576.
[29] F. Virard, S. Cousty, J.P. Cambus, A. Valentin, P. Kemoun, F. Clement, Cold
[1] M. Laroussi, Sterilization of contaminated matter with an atmospheric atmospheric plasma induces a predominantly necrotic cell death via the
pressure plasma, IEEE Trans. Plasma Sci. 24 (1996) 1188–1191. microenvironment, PLoS One 10 (2015) e0133120.
[2] D.B. Graves, The emerging role of reactive oxygen and nitrogen species in [30] J. Schlegel, J. Köritzer, V. Boxhammer, Plasma in cancer treatment, Clin.
redox biology and some implications for plasma applications to medicine Plasma Med. 1 (2013) 2–7.
and biology, J. Phys. D: Appl. Phys. 45 (2012) 263001. [31] S. Salehi, A. Shokri, M.R. Khani, M. Bigdeli, B. Shokri, Investigating effects of
[3] D.B. Graves, Oxy-nitroso shielding burst model of cold atmospheric plasma atmospheric-pressure plasma on the process of wound healing, Biointer-
therapeutics, Clin. Plasma Med. 2 (2014) 38–49. phases 10 (2015) 029504.
[4] N. Kaushik, N. Kumar, C.H. Kim, N.K. Kaushik, E.H. Choi, Dielectric barrier [32] V.N. Vasilets, A. Gutsol, A.B. Shekhter, A. Fridman, Plasma medicine, High
discharge plasma efficiently delivers an apoptotic response in human Energy Chem. 43 (2009) 229–233.
monocytic lymphoma, Plasma Process. Polym. 11 (2014) 1175–1187. [33] A.S. Wu, S. Kalghatgi, D. Dobrynin, R. Sensenig, E. Cerchar, E. Podolsky, et al.,
[5] A. Mai-Prochnow, M. Bradbury, K. Ostrikov, A.B. Murphy, Pseudomonas Porcine intact and wounded skin responses to atmospheric nonthermal
aeruginosa biofilm response and resistance to cold atmospheric pressure plasma, J. Surg. Res. 179 (2013) e1–e12.
plasma is linked to the redox-active molecule phenazine, PLoS One 10 (2015) [34] J. Winter, J.S. Sousa, N. Sadeghi, A. Schmidt-Bleker, S. Reuter, V. Puech, The
e0130373. spatio-temporal distribution of he (23s1) metastable atoms in a mhz-driven
[6] X. Cheng, J. Sherman, W. Murphy, E. Ratovitski, J. Canady, M. Keidar, The helium plasma jet is influenced by the oxygen/nitrogen ratio of the sur-
effect of tuning cold plasma composition on glioblastoma cell viability, PLoS rounding atmosphere, Plasma Sources Sci. Technol. 24 (2015) 25015–25025.
One 9 (2014) e98652. [35] M.A. Lieberman, A.J. Lichtenberg, Principles of plasma discharges and ma-
[7] J.W. Lackmann, S. Schneider, E. Edengeiser, F. Jarzina, S. Brinckmann, terials processing, MRS Bull. 30 (1994) 899–901.
E. Steinborn, et al., Photons and particles emitted from cold atmospheric- [36] K.D. Weltmann, E. Kindel, R. Brandenburg, C. Meyer, R. Bussiahn, C. Wilke,
pressure plasma inactivate bacteria and biomolecules independently and et al., Atmospheric pressure plasma jet for medical therapy: plasma para-
synergistically, J. R. Soc. Interface 10 (2013) 20130591. meters and risk estimation, Contrib. Plasma Phys. 49 (2009) 631–640.
[8] S. Samukawa, M. Hori, S. Rauf, K. Tachibana, P. Bruggeman, G. Kroesen, et al., [37] J. Winter, K. Wende, K. Masur, S. Iseni, M. Dünnbier, M.U. Hammer, et al.,
The 2012 plasma roadmap, J. Phys. D: Appl. Phys. 45 (2012) 253001. Feed gas humidity: a vital parameter affecting a cold atmospheric-pressure
[9] M.D. Ionita, M. Teodorescu, C. Stancu, E.C. Stancu, E.R. Ionita, A. Moldovan, plasma jet and plasma-treated human skin cells, J. Phys. D: Appl. Phys. 46
et al., Surface modification of polymers at atmospheric pressure in expand- (2013) 295401.
ing rf plasmas generated by planar dielectric barrier discharges, J. Optoe- [38] S. Reuter, J. Winter, S. Iseni, S. Peters, A. Schmidt-Bleker, M. Dünnbier, et al.,
lectron. Adv. Mater. 12 (2010) 777–782. Detection of ozone in a mhz argon plasma bullet jet, Plasma Sources Sci.
[10] M.S. Kyi, J. Holton, G.L. Ridgway, Assessment of the efficacy of a low tem- Technol. 21 (2012) 034015.
perature hydrogen peroxide gas plasma sterilization system, J. Hosp. Infect. [39] K.D. Weltmann, T. von Woedtke, Basic requirements for plasma sources in
31 (1995) 275–284. medicine, Eur. J. Appl. Phys. 55 (2011) 13807.
[11] W.A. Rutala, M.F. Gergen, D.J. Weber, Comparative evaluation of the spor- [40] S. Reuter, J. Winter, A. Schmidt-Bleker, H. Tresp, M.U. Hammer, K.-
icidal activity of new low-temperature sterilization technologies: ethylene D. Weltmann, Controlling the ambient air affected reactive species compo-
oxide, 2 plasma sterilization systems, and liquid peracetic acid, Am. J. Infect. sition in the effluent of an argon plasma jet, IEEE Trans. Plasma Sci. 40 (2012)
Control 26 (1998) 393–398. 2788–2794.
[12] N.N. Misra, B.K. Tiwari, Raghavarao KSMS, P.J. Cullen, Nonthermal plasma [41] A. Schmidt-Bleker, J. Winter, S. Iseni, M. Dünnbier, K.D. Weltmann, S. Reuter,
inactivation of food-borne pathogens, Food Eng. Rev. 3 (2011) 159–170. Reactive species output of a plasma jet with a shielding gas device—com-
[13] P. Basaran, N. Basaran-Akgul, L. Oksuz, Elimination of aspergillus parasiticus bination of ftir absorption spectroscopy and gas phase modelling, J. Phys. D:
from nut surface with low pressure cold plasma (lpcp) treatment, Food Mi- Appl. Phys. 47 (2014) 145201.
crobiol. 25 (2008) 626–632. [42] K. Wende, S. Reuter, T. von Woedtke, K.-D. Weltmann, K. Masur, Redox-based
[14] S. Perni, D.W. Liu, G. Shama, M.G. Kong, Cold atmospheric plasma deconta- assay for assessment of biological impact of plasma treatment, Plasma Pro-
mination of the pericarps of fruit, J. Food Prot. 71 (2008) 302–308. cess. Polym. 11 (2014) 655–663.
[15] A.S. Chiper, W. Chen, O. Mejlholm, P. Dalgaard, E. Stamate, Atmospheric [43] T. Adachi, H. Tanaka, S. Nonomura, H. Hara, S. Kondo, M. Hori, Plasma-acti-
pressure plasma produced inside a closed package by a dielectric barrier vated medium induces a549 cell injury via a spiral apoptotic cascade in-
discharge in ar/co2 for bacterial inactivation of biological samples, Plasma volving the mitochondrial-nuclear network, Free Radic. Biol. Med. 79 (2015)
Sources Sci. Technol. 20 (2011) 025008. 28–44.
[16] D. Ziuzina, S. Patil, P.J. Cullen, K.M. Keener, P. Bourke, Atmospheric cold [44] D. Dobrynin, K. Arjunan, A. Fridman, G. Friedman, A.M. Clyne, Direct and
plasma inactivation of escherichia coli in liquid media inside a sealed controllable nitric oxide delivery into biological media and living cells by a
package, J. Appl. Microbiol. 114 (2013) 778–787. pin-to-hole spark discharge (phd) plasma, J. Phys. D: Appl. Phys. 44 (2011)
[17] P. Muranyi, J. Wunderlich, H.C. Langowski, Modification of bacterial struc- 075201.
tures by a low-temperature gas plasma and influence on packaging material, [45] K. Priya Arjunan, A. Morss Clyne, Hydroxyl radical and hydrogen peroxide
J. Appl. Microbiol. 109 (2010) 1875–1885. are primarily responsible for dielectric barrier discharge plasma-induced
[18] K. Duske, L. Jablonowski, I. Koban, R. Matthes, B. Holtfreter, A. Sckell, et al., angiogenesis, Plasma Process. Polym. 8 (2011) 1154–1164.
Cold atmospheric plasma in combination with mechanical treatment im- [46] F. Liu, P. Sun, N. Bai, Y. Tian, H. Zhou, S. Wei, et al., Inactivation of bacteria in
proves osteoblast growth on biofilm covered titanium discs, Biomaterials 52 an aqueous environment by a direct-current, cold-atmospheric-pressure air
(2015) 327–334. plasma microjet, Plasma Process. Polym. 7 (2010) 231–236.
[19] C. Gabler, C. Zietz, R. Gohler, A. Fritsche, T. Lindner, M. Haenle, et al., Eva- [47] M. Golkowski, C. Golkowski, J. Leszczynski, S.R. Plimpton, P. Maslowski,
luation of osseointegration of titanium alloyed implants modified by plasma A. Foltynowicz, et al., Hydrogen-peroxide-enhanced nonthermal plasma ef-
polymerization, Int. J. Mol. Sci. 15 (2014) 2454–2464. fluent for biomedical applications, IEEE Trans. Plasma Sci. 40 (2012)
[20] F. Intranuovo, P. Favia, E. Sardella, C. Ingrosso, M. Nardulli, R. d'Agostino, 1984–1991.
et al., Osteoblast-like cell behavior on plasma deposited micro/nano- [48] T. Shimizu, T. Nosenko, G.E. Morfill, T. Sato, H.-U. Schmidt, T. Urayama,
patterned coatings, Biomacromolecules 12 (2011) 380–387. Characterization of low-temperature microwave plasma treatment with and
[21] H.J. Ahn, K.I. Kim, N.N. Hoan, C.H. Kim, E. Moon, K.S. Choi, et al., Targeting without uv light for disinfection, Plasma Process. Polym. 7 (2010) 288–293.
cancer cells with reactive oxygen and nitrogen species generated by atmo- [49] S. Ikawa, K. Kitano, S. Hamaguchi, Effects of ph on bacterial inactivation in
spheric-pressure air plasma, PLoS One 9 (2014) e86173. aqueous solutions due to low-temperature atmospheric pressure plasma
[22] N. Barekzi, M. Laroussi, Effects of low temperature plasmas on cancer cells, application, Plasma Process. Polym. 7 (2010) 33–42.
Plasma Process. Polym. 10 (2013) 1039–1050. [50] S. Kalghatgi, C.M. Kelly, E. Cerchar, B. Torabi, O. Alekseev, A. Fridman, et al., Ef-
[23] J.W. Chang, S.U. Kang, Y.S. Shin, K.I. Kim, S.J. Seo, S.S. Yang, et al., Non-thermal fects of non-thermal plasma on mammalian cells, PLoS One 6 (2011) e16270.
atmospheric pressure plasma inhibits thyroid papillary cancer cell invasion [51] S. Guo, L.A. Dipietro, Factors affecting wound healing, J. Dent. Res. 89 (2010)
via cytoskeletal modulation, altered mmp-2/-9/upa activity, PLoS One 9 219–229.
(2014) e92198. [52] F.H. Epstein, A.J. Singer, R.A.F. Clark, Cutaneous wound healing, N. Engl. J.
[24] F. Gesbert, L. Larue, Non-thermal plasmas: Novel preventive and curative Med. 341 (1999) 738–746.
therapy against melanomas? Exp. Dermatol. 23 (2014) 716–717. [53] T.A. Wilgus, Immune cells in the healing skin wound: Influential players at
[25] N. Hattori, S. Yamada, K. Torii, S. Takeda, K. Nakamura, H. Tanaka, et al., Ef- each stage of repair, Pharmacol. Res. 58 (2008) 112–116.
fectiveness of plasma treatment on pancreatic cancer cells, Int. J. Oncol. 47 [54] N.D. Evans, R.O. Oreffo, E. Healy, P.J. Thurner, Y.H. Man, Epithelial mechan-
(2015) 1655–1662. obiology, skin wound healing, and the stem cell niche, J. Mech. Behav.
[26] Y. Ma, C.S. Ha, S.W. Hwang, H.J. Lee, G.C. Kim, K.W. Lee, et al., Non-thermal Biomed. Mater. 28 (2013) 397–409.
atmospheric pressure plasma preferentially induces apoptosis in p53-mu- [55] I. Stamenkovic, Extracellular matrix remodelling: the role of matrix me-
tated cancer cells by activating ros stress-response pathways, PLoS One 9 talloproteinases, J. Pathol. 200 (2003) 448–464.
(2014) e91947. [56] K. Rosner, C. Ross, T. Karlsmark, A.A. Petersen, F. Gottrup, G.L. Vejlsgaard,
[27] X. Tan, S. Zhao, Q. Lei, X. Lu, G. He, K. Ostrikov, Single-cell-precision micro- Immunohistochemical characterization of the cutaneous cellular infiltrate in
plasma-induced cancer cell apoptosis, PLoS One 9 (2014) e101299. different areas of chronic leg ulcers, APMIS 103 (1995) 293–299.
[28] F. Utsumi, H. Kajiyama, K. Nakamura, H. Tanaka, M. Mizuno, K. Ishikawa, [57] G.C. Gurtner, S. Werner, Y. Barrandon, M.T. Longaker, Wound repair and
et al., Effect of indirect nonequilibrium atmospheric pressure plasma on anti- regeneration, Nature 453 (2008) 314–321.
26 S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28

[58] S.A. Eming, T. Krieg, J.M. Davidson, Inflammation in wound repair: Molecular [90] E. Dolezalova, P. Lukes, Membrane damage and active but nonculturable
and cellular mechanisms, J. Invest. Dermatol. 127 (2007) 514–525. state in liquid cultures of escherichia coli treated with an atmospheric
[59] B. Mahdavian Delavary, W.M. van der Veer, M. van Egmond, F.B. Niessen, R. pressure plasma jet, Bioelectrochemistry 103 (2015) 7–14.
H. Beelen, Macrophages in skin injury and repair, Immunobiology 216 (2011) [91] G. Daeschlein, M. Napp, S. Lutze, A. Arnold, S. von Podewils, D. Guembel,
753–762. et al., Skin and wound decontamination of multidrug-resistant bacteria by
[60] W.K. Wu, O.P. Llewellyn, D.O. Bates, L.B. Nicholson, A.D. Dick, Il-10 regulation cold atmospheric plasma coagulation, J. Dtsch. Dermatol. Ges. 13 (2015)
of macrophage vegf production is dependent on macrophage polarisation 143–150.
and hypoxia, Immunobiology 215 (2010) 796–803. [92] K. Fricke, I. Koban, H. Tresp, L. Jablonowski, K. Schroder, A. Kramer, et al.,
[61] C.K. Sen, L. Packer, Antioxidant and redox regulation of gene transcription, Atmospheric pressure plasma: A high-performance tool for the efficient
FASEB J. 10 (1996) 709–720. removal of biofilms, PLoS One 7 (2012) e42539.
[62] W. Droge, Free radicals in the physiological control of cell function, Physiol. [93] R. Matthes, I. Koban, C. Bender, K. Masur, E. Kindel, K.-D. Weltmann, et al.,
Rev. 82 (2002) 47–95. Antimicrobial efficacy of an atmospheric pressure plasma jet against biofilms
[63] D. Salvemini, R. Botting, Modulation of platelet function by free radicals and ofpseudomonas aeruginosaandstaphylococcus epidermidis, Plasma Process.
free-radical scavengers, Trends Pharmacol. Sci. 14 (1993) 36–42. Polym. 10 (2013) 161–166.
[64] P. Pignatelli, F.M. Pulcinelli, L. Lenti, P.P. Gazzaniga, F. Violi, Hydrogen per- [94] I. Koban, M.H. Geisel, B. Holtfreter, L. Jablonowski, N.O. Hubner, R. Matthes,
oxide is involved in collagen-induced platelet activation, Blood 91 (1998) et al., Synergistic effects of nonthermal plasma and disinfecting agents
484–490. against dental biofilms in vitro, ISRN Dent. 2013 (2013) 573262.
[65] B.M. Babior, Oxygen-dependent microbial killing by phagocytes, N. Engl. J. [95] G. Daeschlein, M. Napp, S. von Podewils, S. Lutze, S. Emmert, A. Lange, et al.,
Med. 298 (1978) 659–668. In vitro susceptibility of multidrug resistant skin and wound pathogens
[66] T. Finkel, Redox-dependent signal transduction, FEBS Lett. 476 (2000) 52–54. against low temperature atmospheric pressure plasma jet (appj) and di-
[67] P.G. Heyworth, A.R. Cross, J.T. Curnutte, Chronic granulomatous disease, Curr. electric barrier discharge plasma (dbd), Plasma Process. Polym. 11 (2014)
Opin. Immunol. 15 (2003) 578–584. 175–183.
[68] M.H. Kim, W. Liu, D.L. Borjesson, F.R. Curry, L.S. Miller, A.L. Cheung, et al., [96] U. Schnabel, K. Oehmigen, K. Naujox, K. Rackow, U. Krohmann, O. Schmitt,
Dynamics of neutrophil infiltration during cutaneous wound healing and et al., Inactivation of escherichia coli k-12 and enterohemorrhagic escher-
infection using fluorescence imaging, J. Invest. Dermatol. 128 (2008) ichia coli (ehec) by atmospheric pressure plasma, J. Agric. Sci. Appl. 3 (2014)
1812–1820. 81–91.
[69] V. Oliveira-Marques, T. Silva, F. Cunha, G. Covas, H.S. Marinho, F. Antunes, [97] A. Hammann, N.O. Huebner, C. Bender, A. Ekkernkamp, B. Hartmann, P. Hinz,
et al., A quantitative study of the cell-type specific modulation of c-rel by et al., Antiseptic efficacy and tolerance of tissue-tolerable plasma compared
hydrogen peroxide and tnf-alpha, Redox Biol. 1 (2013) 347–352. with two wound antiseptics on artificially bacterially contaminated eyes
[70] P. Ranjan, V. Anathy, P.M. Burch, K. Weirather, J.D. Lambeth, N.H. Heintz, from commercially slaughtered pigs, Skin Pharmacol. Physiol. 23 (2010)
Redox-dependent expression of cyclin d1 and cell proliferation by nox1 in 328–332.
mouse lung epithelial cells, Antioxid. Redox Signal. 8 (2006) 1447–1459. [98] I. Koban, R. Matthes, N.-O. Hübner, A. Welk, P. Meisel, B. Holtfreter, et al.,
[71] C.K. Sen, S. Roy, Redox signals in wound healing, Biochim. Biophys. Acta 1780 Treatment ofcandida albicansbiofilms with low-temperature plasma induced
(2008) 1348–1361. by dielectric barrier discharge and atmospheric pressure plasma jet, New J.
[72] S. Roy, S. Khanna, K. Nallu, T.K. Hunt, C.K. Sen, Dermal wound healing is Phys. 12 (2010) 073039.
subject to redox control, Mol. Ther. 13 (2006) 211–220. [99] G. Daeschlein, S. Scholz, T. von Woedtke, M. Niggemeier, E. Kindel,
[73] B.C. Nwomeh, D.R. Yager, I.K. Cohen, Physiology of the chronic wound, Clin. R. Brandenburg, et al., In vitro killing of clinical fungal strains by low-tem-
Plast. Surg. 25 (1998) 341–356. perature atmospheric-pressure plasma jet, IEEE Trans. Plasma Sci. 39 (2011)
[74] G.S. Lazarus, Definitions and guidelines for assessment of wounds and eva- 815–821.
luation of healing, Arch. Dermatol. 130 (1994) 489–493. [100] I. Koban, N.-O. Hübner, R. Matthes, A. Welk, E. Kindel, K.-D. Weltmann, et al.,
[75] M. Loots, Fibroblasts derived from chronic diabetic ulcers differ in their re- Antiseptic efficacy of selected agents and tissue tolerable plasma (ttp) on c.
sponse to stimulation with egf, igf-i, bfgf and pdgf-ab compared to controls, Albicans biofilms – has the biofilm maturity influence on it? GMS Kranken-
Eur. J. Cell Biol. 81 (2002) 153–160. haushygiene Interdiszip. 4 (2009) 1–8.
[76] P.G. Bowler, Wound pathophysiology, infection and therapeutic options, Ann. [101] G. Daeschlein, S. Scholz, A. Arnold, T. von Woedtke, E. Kindel, M. Niggemeier,
Med. 34 (2002) 419–427. et al., In vitro activity of atmospheric pressure plasma jet (appj) plasma
[77] N.B. Menke, K.R. Ward, T.M. Witten, D.G. Bonchev, R.F. Diegelmann, Impaired against clinical isolates of demodex folliculorum, IEEE Trans. Plasma Sci. 38
wound healing, Clin. Dermatol. 25 (2007) 19–25. (2010) 2969–2973.
[78] R.F. Diegelmann, Excessive neutrophils characterize chronic pressure ulcers, [102] T.K. Hunt, H. Hopf, Z. Hussain, Physiology of wound healing, Adv. Skin Wound
Wound Repair Regen. 11 (2003) 490–495. Care 13 (2000) 6–11.
[79] S.A. Eming, M. Koch, A. Krieger, B. Brachvogel, S. Kreft, L. Bruckner-Tuder- [103] K. Wende, K. Landsberg, U. Lindequist, K.-D. Weltmann, T. von Woedtke,
man, et al., Differential proteomic analysis distinguishes tissue repair bio- Distinctive activity of a nonthermal atmospheric-pressure plasma jet on
marker signatures in wound exudates obtained from normal healing and eukaryotic and prokaryotic cells in a cocultivation approach of keratinocytes
chronic wounds, J Proteome Res. 9 (2010) 4758–4766. and microorganisms, IEEE Trans. Plasma Sci. 38 (2010) 2479–2485.
[80] V. Falanga, Wound healing and its impairment in the diabetic foot, Lancet [104] A. Barton, K. Wende, L. Bundscherer, S. Hasse, A. Schmidt, S. Bekeschus, et al.,
366 (2005) 1736–1743. Non-thermal plasma increases expression of wound healing related genes in
[81] R. Renner, M. Sticherling, R. Ruger, J. Simon, Persistence of bacteria like a keratinocyte cell line, Plasma Med. 3 (2013) 125–136.
pseudomonas aeruginosa in non-healing venous ulcers, Eur. J. Dermatol. 22 [105] A. Schmidt, K. Wende, S. Bekeschus, L. Bundscherer, A. Barton, K. Ottmuller,
(2012) 751–757. et al., Non-thermal plasma treatment is associated with changes in tran-
[82] R.G. Sibbald, P. Coutts, K.Y. Woo, Reduction of bacterial burden and pain in scriptome of human epithelial skin cells, Free Radic. Res. 47 (2013) 577–592.
chronic wounds using a new polyhexamethylene biguanide antimicrobial [106] K. Wende, A. Barton, S. Bekeschus, L. Bundscherer, A. Schmidt, K.-
foam dressing-clinical trial results, Adv. Skin Wound Care 24 (2011) 78–84. D. Weltmann, et al., Proteomic tools to characterize non-thermal plasma
[83] R.E.J. Sladek, T.A. Baede, E. Stoffels, Plasma-needle treatment of substrates effects in eukaryotic cells, Plasma Med. 3 (2013) 81–95.
with respect to wettability and growth of escherichia coli and streptococcus [107] A. Schmidt, S. Dietrich, A. Steuer, K.D. Weltmann, T. von Woedtke, K. Masur,
mutans, IEEE Trans. Plasma Sci. 34 (2006) 1325–1330. et al., Non-thermal plasma activates human keratinocytes by stimulation of
[84] E. Sysolyatina, A. Mukhachev, M. Yurova, M. Grushin, V. Karalnik, antioxidant and phase ii pathways, J. Biol. Chem. 290 (2015) 6731–6750.
A. Petryakov, et al., Role of the charged particles in bacteria inactivation by [108] A. Kramer, J. Lademann, C. Bender, A. Sckell, B. Hartmann, S. Münch, et al.,
plasma of a positive and negative corona in ambient air, Plasma Process. Suitability of tissue tolerable plasmas (ttp) for the management of chronic
Polym. 11 (2014) 315–334. wounds, Clin. Plasma Med. 1 (2013) 11–18.
[85] V. Boxhammer, G.E. Morfill, J.R. Jokipii, T. Shimizu, T. Klämpfl, Y.F. Li, et al., [109] S. Hasse, T. Duong Tran, O. Hahn, S. Kindler, H.R. Metelmann, T. von Woedtke,
Bactericidal action of cold atmospheric plasma in solution, New J. Phys. 14 et al., Induction of proliferation of basal epidermal keratinocytes by cold
(2012) 113042. atmospheric-pressure plasma, Clin. Exp Dermatol (2015), http://dx.doi.org/
[86] K.D. Weltmann, R. Brandenburg, T. von Woedtke, J. Ehlbeck, R. Foest, 10.1111/ced.12735, accepted.
M. Stieber, et al., Antimicrobial treatment of heat sensitive products by [110] S. Bekeschus, Effects of Cold Physical Plasma on Human Leukocytes (dis-
miniaturized atmospheric pressure plasma jets (appjs), J. Phys. D: Appl. Phys. sertation) [Monography], University of Greifswald, Greifswald, 2015.
41 (2008) 194008. [111] V. Brinkmann, U. Reichard, C. Goosmann, B. Fauler, Y. Uhlemann, D.S. Weiss,
[87] I. Koban, B. Holtfreter, N.O. Hubner, R. Matthes, R. Sietmann, E. Kindel, et al., et al., Neutrophil extracellular traps kill bacteria, Science 303 (2004)
Antimicrobial efficacy of non-thermal plasma in comparison to chlorhex- 1532–1535.
idine against dental biofilms on titanium discs in vitro-proof of principle [112] L. Bundscherer, S. Bekeschus, H. Tresp, S. Hasse, S. Reuter, K.-D. Weltmann,
experiment, J. Clin. Periodontol. 38 (2011) 956–965. et al., Viability of human blood leucocytes compared with their respective
[88] N.O. Hubner, R. Matthes, I. Koban, C. Randler, G. Muller, C. Bender, et al., cell lines after plasma treatment, Plasma Med. 3 (2013) 71–80.
Efficacy of chlorhexidine, polihexanide and tissue-tolerable plasma against [113] S. Bekeschus, J. Kolata, A. Muller, A. Kramer, K.-D. Weltmann, B. Broker, et al.,
pseudomonas aeruginosa biofilms grown on polystyrene and silicone ma- Differential viability of eight human blood mononuclear cell subpopulations
terials, Skin Pharmacol. Physiol. 23 (2010) 28–34. after plasma treatment, Plasma Med. 3 (2013) 1–13.
[89] G. Daeschlein, S. Scholz, S. Emmert, S. von Podewils, H. Haase, T. von Woedtke, [114] S. Bekeschus, J. Kolata, C. Winterbourn, A. Kramer, R. Turner, K.D. Weltmann,
et al., Plasma medicine in dermatology: Basic antimicrobial efficacy testing as et al., Hydrogen peroxide: A central player in physical plasma-induced oxi-
prerequisite to clinical plasma therapy, Plasma Med. 2 (2012) 33–69. dative stress in human blood cells, Free Radic. Res. 48 (2014) 542–549.
S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28 27

[115] S. Bekeschus, T. von Woedtke, A. Kramer, K.-D. Weltmann, K. Masur, Cold [141] R. Bussiahn, N. Lembke, R. Gesche, T. von Woedtke, K.-D. Weltmann, Plas-
physical plasma treatment alters redox balance in human immune cells, maquellen für biomedizinische applikationen, Hyg. Med. 38 (2013) 212–216.
Plasma Med. 3 (2013) 267–278. [142] European Union, Directive 2006/25/ec on the minimum health and safety
[116] S. Bekeschus, S. Iseni, S. Reuter, K. Masur, K.-D. Weltmann, Nitrogen shielding requirements regarding the exposure of workers to risks arising from phy-
of an argon plasma jet and its effects on human immune cells, IEEE Trans. sical agents (artificial optical radiation), in: Procedings of the Ninteenth In-
Plasma Sci. 43 (2015) 776–781. dividual Directive Within the Meaning of Article 16(1) of Directive 89/391/
[117] J. Winter, H. Tresp, M.U. Hammer, S. Iseni, S. Kupsch, A. Schmidt-Bleker, et al., eec), 2006, pp. 0038–0059.
Tracking plasma generated h2o2from gas into liquid phase and revealing its [143] S.A. Norberg, W. Tian, E. Johnsen, M.J. Kushner, Atmospheric pressure plasma
dominant impact on human skin cells, J. Phys. D: Appl. Phys. 47 (2014) jets interacting with liquid covered tissue: touching and not-touching the
285401. liquid, J. Phys. D: Appl. Phys. 47 (2014) 475203.
[118] M.A. Loots, E.N. Lamme, J. Zeegelaar, J.R. Mekkes, J.D. Bos, E. Middelkoop, [144] W.V. Gaens, S. Iseni, A. Schmidt-Bleker, K.D. Weltmann, S. Reuter, A. Bogaerts,
Differences in cellular infiltrate and extracellular matrix of chronic diabetic Numerical analysis of the effect of nitrogen and oxygen admixtures on the
and venous ulcers versus acute wounds, J. Invest. Dermatol. 111 (1998) chemistry of an argon plasma jet operating at atmospheric pressure, New J.
850–857. Phys. 17 (2015) 033003.
[119] S. Bekeschus, K. Masur, J. Kolata, K. Wende, A. Schmidt, L. Bundscherer, et al., [145] European Union, Directive 2002/3/ec relating to ozone in ambient air, 2002,
Human mononuclear cell survival and proliferation is modulated by cold pp. 0014–0030.
atmospheric plasma jet, Plasma Process. Polym. 10 (2013) 706–713. [146] J.P. Beck, P. Grennfelt, Estimate of ozone production and destruction over
[120] S. Bekeschus, A. Schmidt, L. Bethge, K. Masur, T. von Woedtke, S. Hasse, et al., northwestern Europe, Atmos. Environ. 28 (1994) 129–140.
Redox stimulation of human thp-1 monocytes in response to cold physical [147] OECD, Guideline for testing of chemicals-genetic toxicology: in vitro mam-
plasma, Oxidative Med. Cell. Longev. 2016 (2016) 5910695. malian cell gene mutation test, 1984, p. 476.
[121] G. Daeschlein, S. Scholz, R. Ahmed, T. von Woedtke, H. Haase, M. Niggemeier, [148] K. Wende, S. Bekeschus, A. Schmidt, L. Jatsch, S. Hasse, K. Masur, Risk as-
et al., Skin decontamination by low-temperature atmospheric pressure sessment of a cold argon plasma jet in respect to its mutagenicity, Mutat. Res.
plasma jet and dielectric barrier discharge plasma, J. Hosp. Infect. 81 (2012) (2015) (Submitted).
177–183. [149] OECD, Guideline for testing of chemicals-genetic toxicology: mammalian
[122] M. Klebes, C. Ulrich, F. Kluschke, A. Patzelt, S. Vandersee, H. Richter, et al., erythrocyte micronucleus test, 1983, p. 474.
Combined antibacterial effects of tissue-tolerable plasma and a modern [150] M. Fenech, A.A. Morley, Measurement of micronuclei in lymphocytes, Mutat.
conventional liquid antiseptic on chronic wound treatment, J. Biophotonics 8 Res. 147 (1985) 29–36.
(2015) 382–391. [151] S. Hasse, O. Hahn, S. Kindler, T. von Woedtke, H.-R. Metelmann, K. Masur,
[123] M. Klebes, J. Lademann, S. Philipp, C. Ulrich, A. Patzelt, M. Ulmer, et al., Effects Atmospheric pressure plasma jet application on human oral mucosa mod-
of tissue-tolerable plasma on psoriasis vulgaris treatment compared to ulates tissue regeneration, Plasma Med. 4 (2014) 117–129.
conventional local treatment: a pilot study, Clin. Plasma Med. 2 (2014) [152] J. Lademann, P.J. Caspers, A. van der Pol, H. Richter, A. Patzelt, L. Zastrow,
22–27. et al., In vivoraman spectroscopy detects increased epidermal antioxidative
[124] M.J. Morykwas, L.C. Argenta, E.I. Shelton-Brown, W. McGuirt, Vacuum-as- potential with topically applied carotenoids, Laser Phys. Lett. 6 (2009) 76–79.
sisted closure: A new method for wound control and treatment: Animal [153] J.W. Fluhr, S. Sassning, O. Lademann, M.E. Darvin, S. Schanzer, A. Kramer,
studies and basic foundation, Ann. Plast. Surg. 38 (1997) 553–562. et al., In vivo skin treatment with tissue-tolerable plasma influences skin
[125] A.R. Badiavas, E.V. Badiavas, Potential benefits of allogeneic bone marrow physiology and antioxidant profile in human stratum corneum, Exp. Der-
mesenchymal stem cells for wound healing, Expert Opin. Biol. Ther. 11 (2011) matol. 21 (2012) 130–134.
1447–1454. [154] J. Lademann, C. Ulrich, A. Patzelt, H. Richter, F. Kluschke, M. Klebes, et al., Risk
[126] C. Shrestha, L. Zhao, K. Chen, H. He, Z. Mo, Enhanced healing of diabetic assessment of the application of tissue-tolerable plasma on human skin, Clin.
wounds by subcutaneous administration of human umbilical cord derived Plasma Med. 1 (2013) 5–10.
stem cells and their conditioned media, Int. J. Endocrinol. 2013 (2013) [155] C. Bender, R. Matthes, E. Kindel, A. Kramer, J. Lademann, K.-D. Weltmann,
592454. et al., The irritation potential of nonthermal atmospheric pressure plasma in
[127] A.L. Sander, D. Henrich, C.M. Muth, I. Marzi, J.H. Barker, J.M. Frank, In vivo the het-cam, Plasma Process. Polym. 7 (2010) 318–326.
effect of hyperbaric oxygen on wound angiogenesis and epithelialization, [156] C. Bender, L.I. Partecke, E. Kindel, F. Doring, J. Lademann, C.D. Heidecke, et al.,
Wound Repair Regen. 17 (2009) 179–184. The modified het-cam as a model for the assessment of the inflammatory
[128] J.H. Barker, D. Kjolseth, M. Kim, J. Frank, I. Bondar, E. Uhl, et al., The hairless response to tissue tolerable plasma, Toxicol. In Vitro 25 (2011) 530–537.
mouse ear: An in vivo model for studying wound neovascularization, Wound [157] T. von Woedtke, H.-R. Metelmann, K.-D. Weltmann, Editorial, Clin. Plasma
Repair Regen. 2 (1994) 138–143. Med. 1 (2013) 1–2.
[129] J.M. Davidson, Animal models for wound repair, Arch. Dermatol. Res. 290 [158] G. Daeschlein, S. Scholz, R. Ahmed, A. Majumdar, T. von Woedtke, H. Haase,
(Suppl.) (1998) S1–S11. et al., Cold plasma is well-tolerated and does not disturb skin barrier or re-
[130] C. Ulrich, F. Kluschke, A. Patzelt, S. Vandersee, V.A. Czaika, H. Richter, et al., duce skin moisture, J. Dtsch. Dermatol. Ges. 10 (2012) 509–515.
Clinical use of cold atmospheric pressure argon plasma in chronic leg ulcers: [159] E. Stoffels, Y. Sakiyama, D.B. Graves, Cold atmospheric plasma: Charged
A pilot study, J Wound Care 24 (196) (2015) 198–200, 202–193. species and their interactions with cells and tissues, IEEE Trans. Plasma Sci.
[131] J. Lademann, C. Ulrich, F. Kluschke, S. Vandersee, K. Weltmann, A. Kramer, et 36 (2008) 1441–1457.
al., Application of tissue-tolerable plasma in dermatology: risk assessment [160] M. Moreau, M.G. Feuilloley, N. Orange, J.L. Brisset, Lethal effect of the gliding
and prospects (oral presentation), in: Proceedings of the Fifth International arc discharges on erwinia spp, J. Appl. Microbiol. 98 (2005) 1039–1046.
Conference on Plasma Medicine, ICPM5, Nara, Japan, 2014. [161] Y.F. Hong, J.G. Kang, H.Y. Lee, H.S. Uhm, E. Moon, Y.H. Park, Sterilization effect
[132] H.R. Metelmann, T. von Woedtke, R. Bussiahn, K.D. Weltmann, M. Rieck, of atmospheric plasma on escherichia coli and bacillus subtilis endospores,
R. Khalili, et al., Experimental recovery of co2-laser skin lesions by plasma Lett. Appl. Microbiol. 48 (2009) 33–37.
stimulation, Am. J. Cosmet. Surg. 29 (2012) 52–56. [162] V. Scholtz, J. Julák, Vz Krí̌ ha, The microbicidal effect of low-temperature plasma
[133] H.-R. Metelmann, T.T. Vu, H.T. Do, T.N.B. Le, T.H.A. Hoang, T.T.T. Phi, et al., Scar generated by corona discharge: Comparison of various microorganisms on an
formation of laser skin lesions after cold atmospheric pressure plasma (cap) agar surface or in aqueous suspension, Plasma Process. Polym. 7 (2010) 237–243.
treatment: A clinical long term observation, Clin. Plasma Med. 1 (2013) 30–35. [163] S. Rupf, A. Lehmann, M. Hannig, B. Schafer, A. Schubert, U. Feldmann, et al.,
[134] S. Vandersee, H. Richter, J. Lademann, M. Beyer, A. Kramer, F. Knorr, et al., Killing of adherent oral microbes by a non-thermal atmospheric plasma jet, J.
Laser scanning microscopy as a means to assess the augmentation of tissue Med. Microbiol. 59 (2010) 206–212.
repair by exposition of wounds to tissue tolerable plasma, Laser Phys. Lett. 11 [164] J. Goree, B. Liu, D. Drake, E. Stoffels, Killing of s. Mutans bacteria using a plasma
(2014) 115701. needle at atmospheric pressure, IEEE Trans. Plasma Sci. 34 (2006) 1317–1324.
[135] K. Wende, P. Williams, J. Dalluge, W.V. Gaens, H. Aboubakr, J. Bischof, et al., [165] J.A. Delben, R.M. Murata, X. Wei, M.L. Castro, W.G. Assuncao, N.R.F.A. da Silva,
Identification of the biologically active liquid chemistry induced by a non- et al., Low-temperature plasma: An effective approach against candida al-
thermal atmospheric pressure plasma jet, Biointerphases 10 (2015) 029518. bicans biofilm, Plasma Med. 4 (2014) 231–244.
[136] O. Lademann, H. Richter, A. Patzelt, A. Alborova, D. Humme, K.D. Weltmann, [166] R.E. Sladek, S.K. Filoche, C.H. Sissons, E. Stoffels, Treatment of streptococcus
et al., Application of a plasma-jet for skin antisepsis: Analysis of the thermal mutans biofilms with a nonthermal atmospheric plasma, Lett. Appl. Micro-
action of the plasma by laser scanning microscopy, Laser Phys. Lett. 7 (2010) biol. 45 (2007) 318–323.
458–462. [167] S.A. Ermolaeva, A.F. Varfolomeev, M.Y. Chernukha, D.S. Yurov, M.M. Vasiliev,
[137] nternational Commission on Non-Ionizing Radiation Protection, Guidelines A.A. Kaminskaya, et al., Bactericidal effects of non-thermal argon plasma in
on limits of exposure to ultraviolet radiation of wavelengths between 180 nm vitro, in biofilms and in the animal model of infected wounds, J. Med. Mi-
and 400 nm (incoherent optical radiation), 0017-9078 Contract No.: 2, 2004. crobiol. 60 (2011) 75–83.
[138] F. Wieringa, A. Van der Meulen, An update on optical radiation safety in [168] E. Kvam, B. Davis, F. Mondello, A.L. Garner, Nonthermal atmospheric plasma
europe, IUVA News 7 (2005) 25–29. rapidly disinfects multidrug-resistant microbes by inducing cell surface da-
[139] D. Deutsche Industrienorm, Strahlungsphysik im optischen bereich und mage, Antimicrob. Agents Chemother. 56 (2012) 2028–2036.
lichttechnik–teil 10: photobiologisch wirksame strahlung, grösen, kurzzie- [169] S.G. Joshi, M. Paff, G. Friedman, G. Fridman, A. Fridman, A.D. Brooks, Control
chen und wirkungsspektrum, DIN, 1996. of methicillin-resistant staphylococcus aureus in planktonic form and bio-
[140] H. Jablonowski, R. Bussiahn, M. Hammer, K.-D. Weltmann, T. von Woedtke, films: A biocidal efficacy study of nonthermal dielectric-barrier discharge
S. Reuter, Impact of plasma jet vacuum ultraviolet radiation on reactive plasma, Am. J. Infect. Control 38 (2010) 293–301.
oxygen species generation in bio-relevant liquids, Phys. Plasmas 22 (2015) [170] P. Sun, Y. Sun, H. Wu, W. Zhu, J.L. Lopez, W. Liu, et al., Atmospheric pressure
122008 (1994-present). cold plasma as an antifungal therapy, Appl. Phys. Lett. 98 (2011) 021501.
28 S. Bekeschus et al. / Clinical Plasma Medicine 4 (2016) 19–28

[171] S. Kalghatgi, G. Friedman, A. Fridman, A.M. Clyne, Endothelial cell prolifera- [183] D. Dobrynin, A. Wu, S. Kalghatgi, S. Park, N. Shainsky, K. Wasko, et al., Live pig
tion is enhanced by low dose non-thermal plasma through fibroblast growth skin tissue and wound toxicity of cold plasma treatment, Plasma Med. 1
factor-2 release, Ann. Biomed. Eng. 38 (2010) 748–757. (2011) 93–108.
[172] P. Brun, S. Pathak, I. Castagliuolo, G. Palu, P. Brun, M. Zuin, et al., Helium [184] G. Collet, E. Robert, A. Lenoir, M. Vandamme, T. Darny, S. Dozias, et al., Plasma
generated cold plasma finely regulates activation of human fibroblast-like jet-induced tissue oxygenation: Potentialities for new therapeutic strategies,
primary cells, PLoS One 9 (2014) e104397. Plasma Sources Sci. Technol. 23 (2014) 012005.
[173] O. Volotskova, M.A. Stepp, M. Keidar, Integrin activation by a cold atmo- [185] G. Fridman, G. Friedman, A. Gutsol, A.B. Shekhter, V.N. Vasilets, A. Fridman,
spheric plasma jet, New J. Phys. 14 (2012) 053019. Applied plasma medicine, Plasma Process. Polym. 5 (2008) 503–533.
[174] M.-H.T. Ngo, J.-D. Liao, P.-L. Shao, C.-C. Weng, C.-Y. Chang, Increased fibroblast [186] V. Boxhammer, Y.F. Li, J. Koritzer, T. Shimizu, T. Maisch, H.M. Thomas, et al.,
cell proliferation and migration using atmospheric n2/ar micro-plasma for Investigation of the mutagenic potential of cold atmospheric plasma at
the stimulated release of fibroblast growth factor-7, Plasma Process. Polym. bactericidal dosages, Mutat. Res. 753 (2013) 23–28.
11 (2014) 80–88. [187] G. Isbary, J. Heinlin, T. Shimizu, J.L. Zimmermann, G. Morfill, H.U. Schmidt,
[175] K.P. Arjunan, G. Friedman, A. Fridman, A.M. Clyne, Non-thermal dielectric et al., Successful and safe use of 2 min cold atmospheric argon plasma in
barrier discharge plasma induces angiogenesis through reactive oxygen chronic wounds: Results of a randomized controlled trial, Br. J. Dermatol. 167
species, J. R. Soc. Interface 9 (2012) 147–157. (2012) 404–410.
[176] S. Arndt, M. Landthaler, J.L. Zimmermann, P. Unger, E. Wacker, T. Shimizu, [188] G. Isbary, G. Morfill, H.U. Schmidt, M. Georgi, K. Ramrath, J. Heinlin, et al., A
et al., Effects of cold atmospheric plasma (cap) on ß-defensins, inflammatory first prospective randomized controlled trial to decrease bacterial load using
cytokines, and apoptosis-related molecules in keratinocytes in vitro and cold atmospheric argon plasma on chronic wounds in patients, Br. J. Der-
in vivo, PLoS One 10 (2014) e0120041. matol. 163 (2010) 78–82.
[177] J. Liebmann, J. Scherer, N. Bibinov, P. Rajasekaran, R. Kovacs, R. Gesche, et al., [189] G. Isbary, W. Stolz, T. Shimizu, R. Monetti, W. Bunk, H.-U. Schmidt, et al., Cold
Biological effects of nitric oxide generated by an atmospheric pressure gas- atmospheric argon plasma treatment may accelerate wound healing in
plasma on human skin cells, Nitric Oxide 24 (2011) 8–16. chronic wounds: Results of an open retrospective randomized controlled
[178] S. Strassenburg, U. Greim, R. Bussiahn, B. Haertel, K. Wende, T. Von Woedtke, study in vivo, Clin. Plasma Med. 1 (2013) 25–30.
et al., Comparison of biological effects on human keratinocytes using differ- [190] J. Heinlin, J.L. Zimmermann, F. Zeman, W. Bunk, G. Isbary, M. Landthaler,
ent plasma treatment regimes, Plasma Med. 3 (2013) 57–69. et al., Randomized placebo-controlled human pilot study of cold atmospheric
[179] S. Arndt, P. Unger, E. Wacker, T. Shimizu, J. Heinlin, Y.F. Li, et al., Cold atmo- argon plasma on skin graft donor sites, Wound Repair Regen. 21 (2013)
spheric plasma (cap) changes gene expression of key molecules of the wound 800–807.
healing machinery and improves wound healing in vitro and in vivo, PLoS [191] G. Isbary, T. Shimizu, J. Zimmermann, J. Heinlin, S. Al-Zaabi, M. Rechfeld,
One 8 (2013) e79325. et al., Randomized placebo-controlled clinical trial showed cold atmospheric
[180] E. Garcia-Alcantara, R. Lopez-Callejas, P.R. Morales-Ramirez, R. Pena-Eguiluz, argon plasma relieved acute pain and accelerated healing in herpes zoster,
R. Fajardo-Munoz, A. Mercado-Cabrera, et al., Accelerated mice skin acute Clin. Plasma Med. 2 (2014) 50–55.
wound healing in vivo by combined treatment of argon and helium plasma [192] F. Brehmer, H.A. Haenssle, G. Daeschlein, R. Ahmed, S. Pfeiffer, A. Gorlitz,
needle, Arch. Med. Res. 44 (2013) 169–177. et al., Alleviation of chronic venous leg ulcers with a hand-held dielectric
[181] M.-H. Ngo Thi, P.-L. Shao, J.-D. Liao, C.-C.K. Lin, H.-K. Yip, Enhancement of barrier discharge plasma generator (plasmaderm((r)) vu-2010): results of a
angiogenesis and epithelialization processes in mice with burn wounds
monocentric, two-armed, open, prospective, randomized and controlled trial
through ros/rns signals generated by non-thermal n2/ar micro-plasma,
(nct01415622), J. Eur. Acad. Dermatol. Venereol. 29 (2015) 148–155.
Plasma Process. Polym. 11 (2014) 1076–1088.
[182] Y. Yu, M. Tan, H. Chen, Z. Wu, L. Xu, J. Li, et al., Non-thermal plasma sup-
presses bacterial colonization on skin wound and promotes wound healing in
mice, J. Huazhong Univ. Sci. Technol. Med. Sci. 31 (2011) 390–394.

You might also like