You are on page 1of 9

968 Review Article DOI: 10.1111/j.1610-0387.2010.07495.

Plasma medicine: possible applications


in dermatology
Julia Heinlin1, Gregor Morfill2, Michael Landthaler1, Wilhelm Stolz3, Georg Isbary3, Julia L. Zimmermann2,
Tetsuji Shimizu2, Sigrid Karrer1
(1) Department of Dermatology, University of Regensburg, Germany
(2) Max Planck Institute for Extraterrestrial Physics, Garching, Germany
(3) Department of Dermatology, Schwabing Hospital, Munich, Germany

JDDG; 2010 • 8:968–976 Submitted: 5.3.2010 | Accepted: 29.5.2010

Keywords Summary
• plasma medicine As a result of both the better understanding of complex plasma phenomena
• plasma and the development of new plasma sources in the past few years, plasma
• cold atmospheric plasma medicine has developed into an innovative field of research showing high
• microbiology potential. While thermal plasmas have long been used in various medical fields
• chronic wounds (for instance for cauterization and sterilization of medical instruments), current
• sterilization research mainly focuses on application of non-thermal plasmas.
• disinfection Experiments show that cold atmospheric plasmas (CAPs) allow efficient, con-
tact-free and painless disinfection, even in microscopic openings, without dam-
aging healthy tissue. Plasmas influence biochemical processes and offer new
possibilities for the selective application of individually designable medically
active substances. In dermatology, new horizons are being opened for wound
healing, tissue regeneration, therapy of skin infections, and probably many
more diseases. First clinical trials show the efficacy and tolerability of plasma in
treating infected chronic wounds. A major task will be the introduction of plas-
ma into clinical medicine and, simultaneously, the further investigation of the
mechanisms of action of plasma at the cellular level.

Introduction devices and in packaging of food stuffs. cell membranes, DNA, lipids and pro-
In physics plasma is considered the The development of diverse, usually teins up to studies on plant, animal and
fourth state of matter next to solids, liq- non-thermal atmospheric-pressure plasma human tissue and finally on patients.
uids and gases. In 1879 the British sources makes its use also in the Without damaging surrounding healthy
chemist and physicist William Crookes (bio)medical field possible. While in the tissue, cold atmospheric-pressure plas-
first described it as “radiant matter “ [1]. past only the thermal properties of plas- mas at room temperature lead to diverse
The term “plasma” itself comes from mas (> 80 °C) were utilized – cauteriza- reactions in tissue. Numerous compo-
Greek (“something molded”) and was in- tion, sterilization of heat-resistant instru- nents of plasma such as e. g reactive oxy-
troduced in 1928 by Irving Langmuir, as ments or for cosmetic, reconstructive gen or nitrogen species, charged parti-
the multicomponent mixture of highly procedures – current research is directed cles, electric fields and even UV light
ionized gases reminded him of blood primarily at the non-thermal effects of (Figure 1) are involved in these effects.
plasma. In technology plasmas have been plasma. Research ranges from investiga- Possible uses include – among others –
established for a very long time – for ex- tion of the fundamental physics, study of very rapid and gentle tissue disinfection
ample in the manufacture of television the optimal, individual plasma composi- via inactivation of diverse pathogens
screens or fluorescent tubes. On the basis tion over the interaction of plasmas with (gram-positive and –negative bacteria,
of their high bactericidal effectiveness prokaryotic and eukaryotic cells, viruses, fungi, viruses, spores and parasites), pre-
plasmas are also used to sterilize medical spores and fungi, cell structures such as cise tissue removal and stimulation of

JDDG | 12˙2010 (Band 8) © The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812
Plasma medicine Review Article 969

Table 1: Comparison of UVR intensities of sunlight and MicroPlaSter ␤®


(microwave power 100 W, main (Ar) gas flow rate 1300 sccm, z 20 mm). Data
refer to an irradiation time of 5 min. Intensity of UV irradiation of sun was
measured in Garching (Munich) and is averaged over one year (Data are
gathered by Bernd Steffes and Dr. Tetsuji Shimizu, Max Planck Institute for
Extraterrestrial Physics, Garching, 2009). Plasma treatment of 1 min with
MicroPlaSter ␤® corresponds to 5 min sun exposure with regard to UVC,
to 1 min with regard to UVB and to 10 s sun exposure with regard to UVA
irradiation.

UVC UVB UVA


(180–280 nm) (280–320 nm) (320–400 nm)
Figure 1: Relevant components of plasma.
Charged particles and density of free radicals Sunlight 1–2.5 µW/cm² 30–50 µW/cm² ~ 600 µW/cm²
(reactive oxygen and nitrogen species) are mainly
responsible for the effects of plasma. MicroPlaSter ␤ 10–16 µW/cm² 40–60 µW/cm² < 100 µW/cm²

UVC: 1 min MicroPlaSter␤ ⬇ 5 min sunlight; UVB: 1 min MicroPlaSter ␤ ⬇


wound healing. Plasmas offer the possi- 1 min sunlight; UVA: 1 min MicroPlaSter ␤ ⬇ 10 s sunlight.
bility of a targeted application of individ-
ually composed medically active sub-
stances without requiring a carrier medium temperature and are thus in equilibrium conducting layer (barrier). The dis-
[2]. Of particular interest is the use of with themselves and their surroundings charges are then not “massive” (and
cold plasmas in hospital hygiene, in the [3], while in non-thermal plasmas – e. g. possibly catastrophic) as in lightning,
treatment of diverse skin and infectious due to microwave discharge – ions and but “gentle” – in many small micro-
diseases, in dentistry and in the cosmetic uncharged molecules are much cooler discharges of about 100 nanoseconds
field. A great advantage of this physical than electrons. These plasmas are “cold” duration – from the barrier to the
method, where according to present because the carrier gas (argon, helium, opposite electrode. On average an
knowledge allergic or toxic reactions are air …) is only slightly ionized (typically almost homogeneous “carpet” of dis-
not expected, is the contact-free, pain- 1 part in 1 billion) and therefore the ions charges results – assuming that the
less, self-sterilizing, non-invasive applica- cool down to room temperature very distances to the opposite electrode are
tion that allows for the treatment of rapidly – in fractions of a second. exactly equal. Typically, the distance
heat-sensitive, inhomogeneous surfaces Relevant parameters of medical plasmas between the plasma device and tissue
and even live tissue. are the electron and ion temperatures is 1 mm.
While bactericidal effects are undis- and density, UV irradiation (Table 1), 2) Indirect plasmas are produced be-
puted, most mechanisms of action of optical and infrared emission, the den- tween two electrodes and then trans-
plasma at a molecular level have largely sity of free radicals, the temperature of ported to the target area by a gas flow
remained unstudied. This review article the neutral gas, the gas composition and [7–9]. The individual discharge can
concentrates on various medical uses of the gas flow. Decisive for the action is the be markedly stronger here (there is no
plasmas with a special focus on derma- flow of active, charged particles (electrons, hindrance by a barrier), the transport
tology. positive and negative ions, e. g. Ar⫹, of the charge carriers (and the pro-
Ag⫺) and uncharged atoms and mole- duced molecules) away from the dis-
Fundamental physics cules (such as O3, OH, H2O2, NO, charge region results simply from the
Plasma denotes a partially ionized gas OH radicals etc.). A great advantage of gas flow and from diffusion. Most de-
that consists to a large extent of charged low-temperature plasmas under atmos- vices of this type produce thin (mm
particles such as ions and electrons, free pheric pressure is the possibility to chem- diameter) plasma jets, larger surfaces
radicals, molecules and also of neutral ically “design” the plasma, i. e. its com- can be treated simultaneously by
atoms. In principle naturally occurring position can be varied depending on the joining many such jets or by multi-
(terrestrial and astrophysical plasmas, desired effect (“chemical cocktail”) [4]. electrode systems. Significantly larger
e. g. the sun, lightning and the polar Low-temperature plasmas at atmos- surfaces can be treated than with di-
lights) and artificially produced plasmas pheric pressure can in principle be classi- rect plasmas. Further, the distance
(e. g. in screens, fluorescent tubes). It is fied into 3 types (Table 2): between the device and the skin is to
estimated that natural plasmas constitute 1) In direct plasmas the tissue/skin a certain degree variable, as the skin is
over 99 % of the visible material of the itself serves as an electrode so that in not needed as a plasma electrode,
universe. It is important to differentiate this form current flows through the significantly simplifying use on the
between hot (thermal) and cold (non- body [5]. A common example of this patient.
thermal or low-temperature) plasma de- is the “dielectric barrier discharge” 3) „Hybrid“ plasmas, also termed
pending on the relative temperatures of device (DBD) [6]. These discharges „barrier coronal discharges“, combine
electrons, ions and neutral gas. In ther- are termed barrier discharges, because both techniques discussed above. They
mal plasmas the latter have the same the electrodes are separated by a non- are produced just as direct plasmas,

© The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812 JDDG | 12˙2010 (Band 8)
970 Review Article Plasma medicine

Table 2: Characteristics of the different atmospheric low-temperature plasmas.

Direct plasmas Indirect plasmas „Hybrid“ plasmas


Dielectric barrier Plasma needle, plasma
Technical examples Barrier coronal discharges
discharge device torch
Mode of production of direct
Production of plasma
Skin or tissue functions plasmas with properties of indirect
Mode of production and between 2 electrodes,
as electrode, current plasmas: Due to grounded electrode
properties transport via gas flow
flows through body net with low electrical resistance no
to target area
current flows through tissue
Gas Air Noble gas/air Air
Distance between device
~ mm ~ mm–cm ~ mm
and treated object
Are produced in the Are produced by mixing Are primarily produced
Reactive species
plasma plasma and air in the plasma
UV radiation Relatively weak Relatively strong Relatively weak
Gas temperature ~ Room temperature Hot at the production site ~ Room temperature
Plasma density on the
High Low Relatively high
treated object

but due to a grounded mesh elec- and the plasma composition and can ria with plasma was reported [12]. In
trode no current flows through tissue therefore vary greatly. For bacterial steril- contrast to conventional methods which
anymore [10]. In contrast to DBD ization, for example, with suitable pa- usually require high temperatures or
systems no air space exists between rameters a short application time of a high concentrations of chemical sub-
the barrier and the opposite electrode few seconds is sufficient. stances such as ethylene oxide, ozone or
(in which the micro-discharges oc- In medical use on patients safety comes chlorine, cold plasma can also be used on
cur) and the opposite electrode must first, so that a risk evaluation of the re- heat-sensitive and chemically reactive
be structured (e. g. mesh). The micro spective plasma device must take place. surfaces. Plasma acts rapidly and very
discharges then occur parallel to the Potential risk factors such as electric cur- effectively and penetrates the smallest
surface of the dielectric barrier, which rent, thermal damage of tissue and the openings and hollow spaces. How
is why these electrodes are also intensity of UV emission as well as opti- plasma actually achieves disinfection or
termed surface micro discharge mal duration and intensity of application sterilization is not yet fully understood.
(SMD)” electrodes. These SMD sys- and gas composition must be checked According to current knowledge both
tems are also independent of the dis- exactly. Lademann et al. [11] in studies physical mechanisms (caused by reactive
tance from the surface to be treated on pig’s ears and tape stripping of volun- species, free radicals, UV photons) and
(within certain limits). teers demonstrated that in treatment biological mechanisms (cellular processes
Universal requirements for the composi- with a CE-certified plasma jet with ar- such as DNA and cell membrane dam-
tion of plasmas do not exist. Most cold gon as carrier gas and optimal treatment age) appear to be responsible for the in-
plasmas at atmospheric pressure are pro- parameters practically no UV light activation of bacteria [13]. UV radiation
duced using helium or argon gas, but reached living skin cells (< 1 % at via energy absorption directly damages
production of plasma e. g. from air or 200 µm skin thickness, maximum UV cellular macromolecules on the one
other mixtures is possible. Limits exist emission in this case at 310 nm, small hand, on the other, DNA proteins and
with regard to electric current, UV radi- bands between 325 and 450 nm), as each lipids are irreparably harmed by oxida-
ation (maximum allowed dose individual of the 15 to 25 corneocyte tive stress.
30 µW/cm², for details see SCCP Euro- layers in human skin absorbs about 25 % Numerous studies using varying treat-
pean Commission Report 0949/05) and of the radiation. Temperature effects also ment parameters prove in vitro the effi-
the production of reactive species (limit were negligible, so that no risk potential cacy of low-temperature plasmas towards
for ozone 50 ppb, see CPSC Consumer is expected for this device in vivo. gram-negative and gram-positive bacte-
Products Safety Commission – report of ria, spores, biofilm-producing bacteria,
09/26/2006, for NO2 2 or 5 ppm and Disinfection and sterilization viruses and fungi. In a phase I study
25 ppm for NO over a time period of of inert materials and vital tissue performed by our working group [7, 14]
8 hours, US National Institute for Occu- It has been known for a long time that in which low-temperature plasma at
pational Safety and Health, NIOH). The ionized gases have biocidal effects, but atmospheric pressure was employed
application time depends on the purpose only in 1996 successful killing of bacte- (MicroPlaSter®, a plasma torch with

JDDG | 12˙2010 (Band 8) © The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812
Plasma medicine Review Article 971

Trials on human or animal skin support


this theory. Short treatment times al-
ready lead to a significant reduction of
the bacterial load, but do not cause
macro- or microscopic alterations in vivo
or ex vivo (Pompl et al. [14]: 95 W,
2.45 GHz, argon as carrier gas, ~ 36 °C,
few eV; Daeschlein et al. [21]: 1–5 kV,
1.5 MHz, argon). Only after a 10-minute
treatment with the FE-DBD device vac-
uolization of keratinocytes in human
skin was detectable histologically.
Various hypotheses for the observed
selectivity of plasma for prokaryotic as
opposed to eukaryotic cells, which are
also supported by other authors, were
summarized by Dobrynin et al. [22]: Eu-
karyotic cells due to a different cell me-
tabolism and higher cell organization are
Figure 2: The plasma torch (MicroPlaSter ␤®; 2.45 GHz, 86 W, Ar 2.2 slm) as an example of an indirect better protected from external stress and
plasma source: Plasma is generated at the 6 electrodes and then transported to the target area via gas
flow. The torch itself has to be cooled by air flow. further have a more favorable surface-
volume ratio, so that they are damaged
only by a distinctly higher dose of
6 electrodes, 110 W microwave output biofilms (produced both by gram-nega- poison.
power, argon gas, Figure 2) a reduction tive and gram-positive bacteria) could be The effect of targeted apoptosis can be
of bacterial load on agar plates after removed in less than 20 s and the growth used in the treatment of malignant cells
2 min by 6 log steps that lasted for at of planktonic bacteria in biofilms could which was demonstrated by Fridman et
least 2 days was achieved. Plasma proved be inhibited within 5 s [17]. Further pos- al. in vitro on ATCC A2058 melanoma
to be effective towards gram-positive and sible results were found with respect to cells (FE-DBD plasma treatment) and
gram-negative bacteria, multiresistant protein removal from surfaces of medical Lee et al. on G361 melanoma cells (he-
bacteria such as MRSA and fungi such as devices with helium-oxygen plasma, lium plasma needle, 13.56 MHz, 15 s at
Candida albicans. Studies of our working probably due to the effects of reactive 4 W, 0.5–5 slm) [23, 24].
group up to now suggest that there is no oxygen species [16]. Further pilot studies – also on other
primary as well as no secondary resist- cancer cells – confirm the apoptotic
ance of the pathogens towards plasma Effects of low-temperature plasmas effect [25–27].
(unpublished data). on mammalian cells
Kamgang-Youbi et al. demonstrated that To study the effects and potential toxici- Low-temperature plasmas and wound
even plasma-treated distilled water ties of cold plasmas on mammalian cells healing
(5 min non-thermal atmospheric-pres- diverse studies have been performed. In Already in 1970 Robson and coworkers
sure plasma with air as carrier gas) has in vitro experiments on fibroblasts, en- recognized that more than 105 colony-
vitro significant antimicrobial effects on dothelial and muscle cells have shown forming units of ␤-hemolytic streptococci,
diverse pathogens and can in the future that effects of low-temperature plasmas Staphylococcus aureus and Pseudomonas
be used in treating contaminated materi- are dose- and time-dependent [18]. aeruginosa on a wound suffice to disturb
als/ tissue [15]. The proof that the infec- While a longer treatment duration or wound healing [28]. Wound healing is
tiosity of prions is reduced significantly greater treatment intensity (DBD plasma, also delayed if more than four different
must still be provided, even though a re- 0.1 W/cm², > 60 s) result in apoptosis or bacteria species are found on a wound
duction was shown in prion-protein necrosis of the cells, shorter treatment [19, 28]. Resistant pathogens, especially
models [16]. Low-temperature plasmas time (30 s) or lower intensity lead to a re- methicillin-resistant Staphylococcus aureus
also make a dissolution or removal of versible loss of cell adhesion, temporary strains (MRSA), which can become a
biofilms, often found on catheters, med- increase in cell membrane permeability, global threat, are occurring with increasing
ical implants and teeth, possible. an inhibition of migration or a stimula- frequency in chronic wounds [30, 31].
Biofilms are three-dimensional accumu- tion of cell proliferation (probably due to Studies on cell cultures prove that a
lations of microorganisms that adhere to growth factors such as e. g. FGF2 [19]). plasma treatment can influence wound
a surface and are enclosed by polymeric For plasma-induced cell apoptosis or also healing not only by a reduction of bacte-
substances and can, for example, effec- cell proliferation most likely reactive rial colonization but also by direct effects
tively protect yeasts from attacks out of species, such as oxygen and nitrogen on epidermal and dermal cells [19, 32].
the environment or the immune system. species, are responsible. An interim analysis of the worldwide
This markedly increases resistance to- Low doses that would already be suffi- first randomized clinical trial with
wards antifungal agents. Lee et al., cient to kill bacteria effectively are harm- MicroPlaSter® (parameters see above) in
among others, were able to show that less for human and animal cells [14, 20]. which in the meantime already over

© The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812 JDDG | 12˙2010 (Band 8)
972 Review Article Plasma medicine

150 patients with chronic infected


wounds received low-temperature argon
plasma once daily over 2 to 5 minutes as
an add-on therapy demonstrated in
291 treatments of 36 patients – inde-
pendent of the species of bacteria includ-
ing MRSA – a highly significant reduc-
tion of bacterial load (34 %, p < 10–6) in
wounds treated with plasma in compari-
son to the control wounds without
plasma [33]. Side effects did not occur at
any time, treatment was well-tolerated
by the patients (Figures 3a, b, c). In an
uncontrolled study on 48 patients with
diabetic foot syndrome Fetykov et al. us-
ing the “Plasmafon”, a low-temperature
plasma source with a large share of UV
radiation, double as rapid complete
wound healing and pain reduction
within 5 days in comparison to the con-
trol group [34].
Of additional benefit to wound healing
and regenerative processes are elevated
concentrations of nitric oxide (NO)
which can be generated exogenously by
plasma [35, 36]. Among others vasodila-
tion and normalized microcirculation,
direct bactericidal effects, improvement
of nerve conduction velocity, stimulation
of fibroblasts and vessel growth are
attributed to the induction of cytokines
and growth factors. In animal models
after application of NO generated exoge-
nously in plasma wounds healed on aver-
age about 24.6 % more rapidly than in
the control group. In an uncontrolled
study on 65 diabetes patients with puru-
lent or necrotic wounds on the legs the
treatment with the “Plazon”, a plasma
device where hot plasma with high NO
concentrations is very rapidly cooled to
20–40 °C, leads to accelerated wound
healing [37]. This fact was confirmed by
Lipatov et al. on 40 patients [36]. De-
spite the fact that the “Plazon” system
has been in medical use for 9 years on
thousands of patients, no controlled,
randomized studies exist yet.

Cold plasmas in atopic dermatitis and


other pruritic diseases
Skin colonization with facultatively
pathogenic microorganisms can trigger
various skin diseases including atopic
dermatitis. To verify this effect of
cold plasmas on bacterial colonization
Daeschlein and coworkers treated a
Figure 3: A 61-year-old patient with venous ulcers: wounds before plasma treatment (a), after 7 (b)
and after 11 treatments (c). For the daily therapy of 2 min MicroPlaSter® was used. At the beginning
patient with pronounced Staphylococcus
of plasma treatment Klebsiella oxytoca and Enterobacter cloacae were detectable, after 11th treatment aureus colonization for 3 minutes with
(23 days later) swabs were sterile. a low-temperature atmospheric-pressure

JDDG | 12˙2010 (Band 8) © The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812
Plasma medicine Review Article 973

pain, bleeding, perforation, strictures,


disturbances in swallowing, the develop-
ment of gas emphysema of the mucosa,
neuromuscular stimulation and even gas
explosions have been reported [48].
More recent studies show that even low-
temperature plasmas accelerate blood co-
agulation [49], both in vitro and in vivo
in animal models. After treatment with
FE-DBD significant alterations in the
protein composition and coagulation
factors in blood, even in anticoagulated
patients and patients with hemophilia
were observed. The detailed biochemical
basis for accelerated coagulation is still
largely unknown. It is known that the
natural blood coagulation is triggered by
selective effects on certain blood pro-
teins, but that the release of calcium ions
or pH changes are not of significance
Figure 4: Treatment of a forearm of a patient with hepatogenic pruritus with argon plasma and that the effect is independent of the
(MicroPlaSter ␤®). Therapy was also performed daily over 2 min.
gas temperature or the temperature on
the surface of blood [50].
plasma jet (1–5 kV; 1.5 MHz, argon a restructuring of dermal architecture
gas), which led to a selective eradication that can be confirmed histologically Further applications
of this pathogen, while the physiological [40–42]. In addition to wrinkle treat- A further possible indication in derma-
skin flora with Staphylococcus epidermidis ment the method can also be used in the tology is the treatment of cutaneous
and Staphylococcus haemolyticus remained therapy of, among others, actinic leishmaniasis. Fridman and colleagues
undamaged and was mobilized to the keratoses, seborrheic keratoses, viral demonstrated in a cell culture a success-
skin surface from deeper skin layers papillomas, scars and sun-damaged skin ful 100 % inactivation of Leishmania
(observable by confocal scanning laser including pigmentary disturbance or in major promastigotes within 20 s, while
microscopy) [38]. Mertens et al. also combination with aesthetic-surgical pro- for inactivation of human macrophages
demonstrated a reduction of Staphylococcus cedures. During treatment local or in contrast a two-minute treatment was
aureus by more than tenfold within systemic anesthesia is required. Several required to inactivate 20–30 % [51].
2 days in a patient with atopic dermatitis studies confirm the method’s success Many bacterial skin diseases such as
(with a direct DBD device at a frequency with a reduction of wrinkles by up to impetigo contagiosa, folliculitis or ec-
of 90–700 Hz at 3–7.8 kV). Further, 50 % [43, 44]. thyma, fungal infections such as tinea
distinct improvement of erythema and a pedis or even viral diseases might profit
reduction of pruritus for hours were Blood coagulation from a treatment with cold plasma in the
observed [39]. Local application of high-temperature future. The ability to penetrate textiles
Possibly low-pressure plasmas will in the plasma for hemostasis and for sclerosing (e. g. socks) makes the use of plasma e. g.
future represent therapy options for the angiodysplasias and ablation of tumors in the prevention or therapy of tinea
treatment of diverse pruritic disorder, has been used since about 30 years, e. g. pedis an interesting alternative. Besides
even though no controlled studies have in the form of the argon plasma coagula- large devices for clinics and medical
yet been published on this (Figure 4) and tor (APC) [45, 46] in many surgical dis- offices small devices for home use are
the exact pathomechanism or mecha- ciplines including endoscopic proce- under development. A further revolution
nism of action are still unknown. dures (40–75 W in the MHz range, might also take place in (hospital)
1–10 l gas flow rate, ~ 120 °C, ~ 3 eV). hygiene: The HandPlaSter®, a low-
Cosmetic applications The effect of ionized gas applied without temperature plasma device using barrier
In 2005 the UF Food and Drug Admin- tissue contact is thermal and is based on coronal discharge (BCD) technology
istration licensed plasma skin regeneration protein denaturation and desiccation. In (ⱕ 0.5 W/cm², 18 kVpp, 12.5 kHz) that
technology (PSR) for skin rejuvenation dermatology the technique was first em- was developed for potential use for hand
and for treatment of wrinkles. ployed in 1997 by Katsch et al. for the disinfection allows for in vitro a reduc-
Here a hot, but rapidly cooling plasma, treatment of intraepithelial disorders tion of the bacterial load by over 5 log
that is produced by a radiofrequency (e. g. actinic keratoses, condylomata steps within few seconds (< 10 s) [10].
plasma jet with nitrogen as carrier gas, is acuminata etc) [47]. The depth of pene- Here even multiresistant pathogens such
employed. The heat application induces tration is only 2–3 mm, making the as MRSA are effectively destroyed. This
controlled thermal damage in the skin method relatively free of side effects. new form of (hand) disinfection has
which results in the new production of Nevertheless, depending on site of appli- some advantages over conventional liq-
collagen, a reduction of elastic fibers and cation diverse complications such as uid disinfectants, which must be applied

© The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812 JDDG | 12˙2010 (Band 8)
974 Review Article Plasma medicine

for several minutes and irritate the skin, MRSA: methicillin-resistant Staphylococ- 5 Fridman G, Friedman G, Gutsol A,
while at the same time remaining dis- cus aureus Shekhter AB, Vasilets VN, Fridman A.
tinctly below WHO limits for UV, toxic PSR: plasma skin regeneration Applied Plasma Medicine. Plasma Pro-
gases and electric current [10]. Naturally, SMD: surface micro discharge cess Polym 2008; 5: 503–33.
not only clinics but also public establish- SSCP: Scientific Committee on Consumer 6 Fridman G, Brooks AD, Balasubramanian
ments, nursing homes etc. could profit. Products M, Fridman A, Gutsol A, Vasilets VN,
Cold plasmas may also find applications Ayan H, Friedman G. Comparison of
in dentistry in the future. Due to the direct and indirect effects of non-ther-
ability to enter microscopically small mal atmospheric-pressure plasma on
openings, theoretically therapeutic meas- bacteria. Plasma Process Polym 2007;
ures against periodontitis, chronic gin- 4: 370–5.
givitis and in the treatment of infected 7 Shimizu T, Steffes B, Pompl R,
root canals are possible. Clinical evidence Jamitzky F, Bunk W, Ramrath K,
for this must still be gathered. Further, in Georgi M, Stolz W, Schmidt HU,
vivo the combination with H2O2 showed Urayama T, Fujii S, Morfill GE. Cha-
an increase in the bleaching effect as well racterization of microwave plasma
as protein removal on the surface of torch for decontamination. Plasma
plasma-treated teeth [52, 53]. Process Polym 2008; 5: 577–82.
Julia Heinlin 8 Sladek RE, Stoffels E. Deactivation of
Conclusions Escherichia coli by the plasma needle.
Thanks to interdisciplinary cooperation J Phys D: Applied Physics 2005; 38:
in medicine, physics, chemistry, biology 1716–21.
Correspondence to 9 Cao Z, Walsh JL, Kong MG. Atmos-
and microbiology plasma medicine has
developed into an innovative and dy- pheric plasma jet array in parallel elec-
namic field of research in recent years. tric and gas flow fields for three-dimen-
Even if many questions remain unan- sional surface treatment. Appl Phys
swered – especially the mechanism of in- Lett 2009; 94: 021501.
teraction between plasma and living 10 Morfill GE, Shimizu T, Steffes B,
cells/ tissues and the optimization of Schmidt HU. Nosocomial infections –
plasma composition depending on the a new approach towards preventive me-
desired effect – studies to date illustrate dicine using plasmas. New Journal of
the great potential of plasma medicine. Physics 2009; 11: 115019.
Cold plasmas will surely be available in 11 Lademann J, Richter H, Alborova A,
the near future to an increasing extent Humme D, Patzelt A, Kramer A, Weltmann
both for therapy and prevention of Prof. Dr. Sigrid Karrer KD, Hartmann B, Ottomann C, Fluhr
diverse, particularly infectious diseases. Department of Dermatology JW, Hinz P, Hubner G, Lademann O.
There does exist a vision of a break- University of Regensburg Risk assessment of the application of a
through in medicine comparable to the Franz-Josef-Strauß-Allee 11 plasma jet in dermatology. J Biomed
introduction of antibiotics. The superi- D-93053 Regensburg, Germany Opt 2009; 14: 054025.
ority of plasmas in comparison to previ- Tel.: +49-941-944-9656 12 Laroussi M. Sterilization of Contami-
ous medical standards remains to be Fax: +49-941-944-9657 nated Matter with an Atmospheric
clarified, especially in economic terms. E-mail: Pressure Plasma. IEEE Trans Plasma Sci
sigrid.karrer@klinik.uni-regensburg.de 1996; 24: 1188–91.
13 Kong MG, Kroesen J, Morfill G,
Conflicts of interest References Nosenko T, Shimizu T, van Dijk J,
Some members of our working group 1 Crookes W. On Radiant Matter Spec- Zimmermann JL. Plasma medicine: an
hold patents on diverse plasma devices troscopy: A New Method of Spectrum introductory review. New Journal of
and techniques. A phase II study on the Analysis. Proc Roy Soc 1883; 35: Physics 2009; 11: 115012.
treatment of infected wounds with cold 262–71. 14 Pompl R, Shimizu T, Schmidt HU,
atmospheric plasma (MicroPlaSter®) is 2 Morfill G, Kong MG, Zimmermann Bunk W, Jamitzky F, Steffes B, Ramrath
being financed by the Max Planck Insti- JL. Focus on Plasma Medicine. New K, Peters B, Stolz W, Urayama T,
tute for Extraterrestrial Physics, Munich, Journal of Physics 2009; 11: 115011. Ramasamy R, Fujii S, Morfill GE.
Germany. <<< 3 Fridman A, Chirokov A, Gutsol A. Efficiency and medical compatibility of
Non-thermal atmospheric pressure di- low-temperature plasma sterilization.
Abbreviations scharges. J Phys D: Applied Physics 6th International Conference on
APC: argon plasma coagulator 2005; 38: R1–R24. Reactive Plasmas. Matsushima, Japan,
CAP: cold atmospheric plasma 4 Nosenko T, Shimizu T, Morfill GE. 2006.
DBD: dielectric barrier discharge Designing plasmas for chronic wound 15 Kamgang-Youbi G, Herry JM,
FE-DBD: floating-electrode dielectric disinfection. New Journal of Physics Meylheuc T, Brisset JL, Bellon-
barrier discharge 2009; 11: 115013. Fontaine MN, Doubla A, Naitali M.

JDDG | 12˙2010 (Band 8) © The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812
Plasma medicine Review Article 975

Microbial inactivation using plasma- 25 Kim D, Gweon B, Kim DB, Choe W, oxide. Bulletin of Experimental Biology
activated water obtained by gliding el- Shin JH. A feasibility study for the and Medicine 1998; 126: 829–34.
ectric discharges. Lett Appl Microbiol. cancer therapy using cold plasma. 13th 36 Lipatov KV, Sopromadze MA, Shekhter
2009; 48: 13–8. International Conference on Biomedi- AB, Emel’ianov A, Grachev SV. [Use of
16 Deng X, Shi JJ, Kong MG. Protein cal Engineering. Singapore: ICBME, gas flow with nitrogen oxide (NO-
destruction by a helium atmospheric 2008: 355–57. therapy) in combined treatment of pu-
pressure glow discharge: Capability and 26 Kim G, Lee H, Shon C. The effect of a rulent wounds]. Khirurgiia (Mosk)
mechanisms. J Appl Phys 2007; 101: micro plasma on melanoma (G361) 2002: 41–3.
074701. cancer cells. J Korean Physical Society 37 Shulutko AM, Antropova NV, Kriuger
17 Lee MH, Park BJ, Jin SC, Kim D, Han 2009; 54: 628–32. Iu A. [NO-therapy in the treatment of
I, Kim J, Hyun SO, Chung KH, Park 27 Zang X, Li M, Zhou R, Feng K, Yang purulent and necrotic lesions of lower
JC. Removal and sterilization of S. Ablation of liver cancer cells in vitro extremities in diabetic patients]. Khir-
biofilms and planktonic bacteria by by a plasma needle. Appl Phys Lett urgiia (Mosk) 2004: 43-6.
microwave-induced argon plasma at at- 2008; 93: 021502. 38 Daeschlein G, Darm K, Niggemeier M,
mospheric pressure. New Journal of 28 Robson MC, Heggers JP. Delayed Majunke S, von Woedtke T, Kindel E,
Physics 2009; 11: 115022. wound closure based on bacterial counts. Weltmann KD, Juenger M. Selective
18 Shashurin A, Keidar M, Bronnikov S, J Surg Oncol 1970; 2: 379–83. antistaphylococcal activity of atmos-
Jurjus RA, Stepp MA. Living tissue un- 29 Trengove NJ, Stacey MC, McGechie pheric pressure plasma jet (APPJ) on
der treatment of cold plasma atmos- DF, Mata S. Qualitative bacteriology human skin. Second International
pheric jet. Appl Phys Lett 2008; 93: and leg ulcer healing. J Wound Care Conference on Plasma Medicine. San
181501. 1996; 5: 277–80. Antonio, Texas, USA, 2009.
19 Kalghatgi S, Fridman A, Friedman G, 30 Grundmann H, Aires-de-Sousa M, 39 Mertens N, Helmke A, Goppold A,
Clyne AM. Non-thermal plasma Boyce J, Tiemersma E. Emergence and Emmert S, Kaemling A, Wandke D,
treatment enhances proliferation of resurgence of meticillin-resistant Sta- Vioel W. Low temperature plasma tre-
endothelial cells. Second International phylococcus aureus as a public-health atment of human tissue. Second Inter-
Conference on Plasma Medicine. San threat. Lancet 2006; 368: 874–85. national Conference on Plasma Medi-
Antonio, Texas, USA, 2009. 31 Klein E, Smith DL, Laxminarayan R. cine. San Antonio, Texas, USA, 2009.
20 Sosnin EA, Stoffels E, Erofeev MV, Hospitalizations and deaths caused 40 Bogle MA, Arndt KA, Dover JS. Eva-
Kieft IE, Kunts SE. The effects of UV by methicillin-resistant Staphylococcus luation of plasma skin regeneration
irradiation and gas plasma treatment on aureus, United States, 1999–2005. technology in low-energy full-facial
living mammalian cells and bacteria: a Emerg Infect Dis 2007; 13: 1840–6. rejuvenation. Arch Dermatol 2007;
comparative approach. IEEE Transact 32 Wende K, Landsberg K, Lindequist U, 143: 168-74.
Plasma Sci 2004; 32: 1544–50. Weltmann KD, v. Woedtke T. Microor- 41 Fitzpatrick R, Bernstein E, Iyer S,
21 Daeschlein G, Darm K, Majunke S, ganisms, human cells and cold atmos- Brown D, Andrews P, Penny K. A
Kindel E, Weltmann KD, Juenger M. pheric plasma – looking for an intersec- histopathologic evaluation of the
In vivo monitoring of atmospheric tion. 2nd International Workshop on Plasma Skin Regeneration System
pressure plasma jet (APPJ) skin therapy Plasma-Tissue Interactions. Greifswald, (PSR) versus a standard carbon dioxide
by confocal laserscan microscopy Germany, 2009. resurfacing laser in an animal model.
(CLSM). Second International Confe- 33 Isbary G, Morfill G, Schmidt HU, Lasers Surg Med 2008; 40: 93–9.
rence on Plasma Medicine. San Antonio, Georgi M, Ramrath K, Heinlin J, 42 Foster KW, Moy RL, Fincher EF.
Texas, USA, 2009. Karrer S, Landthaler M, Shimizu T, Advances in plasma skin regeneration.
22 Dobrynin D, Fridman G, Friedman G, Steffes B, Bunk W, Monetti R, J Cosmet Dermatol 2008; 7: 169–79.
Fridman A. Physical and biological me- Zimmermann JL, Pompl R, Stolz W. A 43 Kilmer S, Semchyshyn N, Shah G,
chanisms of direct plasma interaction first prospective randomized controlled Fitzpatrick R. A pilot study on the use
with living tissue. New Journal of Phy- trial to decrease bacterial load using of a plasma skin regeneration device
sics 2009; 11: 115020. cold atmospheric argon plasma on (Portrait PSR3) in full facial rejuvena-
23 Lee HJ, Shon CH, Kim YS, Kim S, chronic wounds in patients. Br J Der- tion procedures. Lasers Med Sci 2007;
Kim GC, Kong MG. Degradation of matol 2010 Mar 5. [Epub ahead of 22: 101–9.
adhesion molecules of G361 melanoma print]. 44 Kilmer S, Fitzpatrick R, Bernstein E,
cells by a non-thermal atmospheric 34 Fetykov AI, Avdeeva EA, Fulton J, Brown D. Long term follow-up on the
pressure microplasma. New Journal of Ferrel J, Gotsev VA, Galov AA. The use of plasma skin regeneration (PSR)
Physics. 2009; 11: 115026. effectiveness of cold plasma treatment in full facial rejuvenation procedures.
24 Fridman G, Shereshevsky A, Jost M, of diabetic feet syndrome, complicated Lasers Surg Med 2005; 36: 22.
Brooks A, Fridman A, Gutsol A, by purulonecrotic process. Second In- 45 Morrison JCF. Electrosurgical method
Vasilets V, Friedman G. Floating elec- ternational Conference on Plasma. San and apparatus for initiating an electrical
trode dielectric barrier discharge Antonio, Texas, USA, 2009. discharge in an inert gas flow. US
plasma in air promoting apoptotic 35 Shekhter AB, Kabisov RK, Pekshev AV, Patent. 1977; 4,040,426.
behavior in melanoma skin cancer cell Kozlov NP, Perov YL. Experimental 46 Ginsberg GG, Barkun AN, Bosco JJ,
lines. Plasma Chem Plasma Process and clinical validation of plasmadyna- Burdick JS, Isenberg GA, Nakao NL,
2007; 27: 163–76. mic therapy of wounds with nitric Petersen BT, Silverman WB, Slivka A,

© The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812 JDDG | 12˙2010 (Band 8)
976 Review Article Plasma medicine

Kelsey PB. The argon plasma coagula- 49 Fridman G, Peddinghaus M, Ayan H, non-thermal atmospheric pressure
tor: February 2002. Gastrointest En- Fridman A, Balasubramanian M, plasma. 37th AIAA Plasmadynamics
dosc 2002; 55: 807–10. Gutsol A, Brooks A, Friedman G. Blood and Lasers Conference. San Francisco,
47 Katsch J, Müller RPA, Mailänder W. coagulation and living tissue sterilization California, USA, 2006.
Argon-Plasma-Koagulation (APC) in by floating-electrode dielectric barrier 52 Sun P, Wang R, Tong G, Zhang J, Fang
der Dermatologie – Eine Standortbe- discharge in air. Plasma Chem Plasma J. Teeth whitening with dental gel assi-
stimmung. VOD Dialog Beilage in Der Process 2006; 26: 425–42. sted by an atmospheric pressure non-
Deutsche Dermatologe 1997; 45: 2–6. 50 Fridman A. Non-thermal plasma assi- thermal plasma. Second International
48 Manner H, Enderle MD, Pech O, May sted blood coagulation. Cambridge Conference on Plasma Medicine. San
A, Plum N, Riemann JF, Ell C, University Press, New York, 2008. Antonio, Texas, USA, 2009.
Eickhoff A. Second-generation argon 51 Fridman G, Shereshevsky A, Pedding- 53 Lee HW, Kim GJ, Kim JM, Park JK,
plasma coagulation: two-center experi- haus M, Gutsol A, Vasilets V, Brooks A, Lee JK, Kim GC. Tooth bleaching
ence with 600 patients. J Gastroenterol Balasubramanian M, Friedman G, with nonthermal atmospheric pressure
Hepatol 2008; 23: 872–8. Fridman A. Bio-medical applications of plasma. J Endod 2009; 35: 587–91.

JDDG | 12˙2010 (Band 8) © The Authors • Journal compilation © Blackwell Verlag GmbH, Berlin • JDDG • 1610-0379/2010/0812

You might also like