You are on page 1of 12

Clin Oral Invest (2015) 19:97107

DOI 10.1007/s00784-014-1224-3

ORIGINAL ARTICLE

In situ antimicrobial activity on oral biofilm: essential


oils vs. 0.2 % chlorhexidine
Victor Quintas & Isabel Prada-Lpez &
Juan Carlos Prados-Frutos & Inmaculada Toms

Received: 8 July 2013 / Accepted: 3 March 2014 / Published online: 1 April 2014
# Springer-Verlag Berlin Heidelberg 2014

Abstract clorhexidine such as a preoperative rinse, in periodontal


Objectives This study aims to evaluate the in situ antibacterial procedures or post-treatment applications.
activity of a mouthwash containing essential oils (M-EO) on
undisturbed de novo plaque-like biofilm (PL-biofilm) up to Keywords Essential oil . Chlorhexidine . Mouthwash .
7 h after its application. Substantivity . Plaque-like biofilm . Laser scanning confocal
Patients and methods An appliance was designed to hold six microscopy
glass disks on the buccal sides of the lower teeth, allowing PL-
biofilm growth. Fifteen healthy volunteers wore the appliance
for 48 h and then performed a M-EO. Disks were removed Introduction
after 30 s and at 1, 3, 5, and 7 h later. After a washout period,
the same procedure was repeated with a M-WATER and a Nowadays, Listerine is the most popular combination of
M-0.2 % chlorhexidine. After PL-biofilm vital staining, sam- essential oils [1] and represents the oldest antigingivitis and
ples were analyzed using a confocal laser scanning antiplaque agent used clinically in dentistry. It is considered
microscope. safe and effective by the Committee of Experts in Oral Health
Results At 30 s after M-EO, the levels of bacterial vitality of the FDA [2]. Listerine contains a fixed combination of
were 1.18 %, significantly lower than that of the basal sample four essential oils (EO) as the active ingredients (thymol
(p<0.001). After 7 h, the antibacterial effect of essential oils 0.064 %, eucalyptol 0.092 %, methyl salicylate 0.060 %,
was still patent with a 47.86 % difference in bacterial vitality menthol 0.042 %). EO kill microorganisms by disrupting their
compared to the basal sample (p<0.001). cell walls and inhibiting their enzymatic activity. They prevent
Conclusion A single M-EO presents high antibacterial imme- bacterial aggregation, slow down bacterial multiplication, and
diate activity and penetration capacity in situ and a extract endotoxins [3].
substantivity which lasts for at least 7 h after its application Recognizing that biofilm bacteria may be ten to 1,000
over de novo biofilm. These results were better than those times more resistant to antimicrobial agents than planktonic
observed with 0.2 % chlorhexidine under the same conditions. cells [4], a more predictive assessment of mouthwash efficacy
Clinical relevance A single M-EO is an effective measure may be better achieved with biofilm tests. Studies have been
against the de novo biofilm, presenting a good alternative to performed on the activity of essential oils on oral biofilm, both
in vitro and in situ. The former, using artificial models, have
helped us gain a better understanding of oral biofilms in spite
of not being predictive of clinical activity [46], as they
V. Quintas : I. Prada-Lpez : I. Toms (*)
involve a limited number of species and they are conducted
Oral Sciences Research Group, School of Medicine and Dentistry,
Santiago de Compostela University, C./ Entrerrios s/n, under conditions which do not reflect the physiological oral
15872 Santiago de Compostela, Spain status [79]. For this reason, several authors have stated that
e-mail: inmaculada.tomas@usc.es results obtained from this type of study must be carefully
interpreted [7, 10, 11].
J. C. Prados-Frutos
Department of Stomatology, Faculty of Health Sciences, Rey Juan With respect to in situ studies, they have greater value
Carlos University, Madrid, Spain when establishing the antiseptic efficacy of several
98 Clin Oral Invest (2015) 19:97107

mouthwashes since their activity is tested under in vivo clin- There are few studies in the literature in which the effects of
ical conditions [1216]. In this type of study, differences have essential oils on in situ undisturbed PL-biofilm have been
been found between those performed on disturbed dental measured by applying CLSM together with bacterial vitality
plaque [1214] and those performed on undisturbed dental techniques [16]. The aim of the present study was to evaluate
plaque [15, 16]. In the first type, the plaque is analyzed after the in situ antibacterial activity of an essential oil mouthwash
being removed from the dental surface [1214]. Due to this, it on undisturbed de novo PL-biofilm up to 7 h after its appli-
is not possible to assess either the original architecture of the cation using CLSM and a dual-stain fluorescence solution.
plaque or the penetration power of the antibacterial agent.
Therefore, methodologies which permit biofilm formation
under real clinical conditions are needed so that plaque dis- Patients and methods
turbance is not necessary for analysis. As a consequence,
several types of removable devices capable of holding multi- This was a randomized, double-blind, crossover study of the
ple sorts of substratum have been devised. These results in a antibacterial efficacy of essential oils on an in situ model of
biofilm which is presumably similar to dental plaque, gener- PL-biofilm growth.
ated under similar conditions and set over an artificial sub-
stratum; this has been called a plaque-like biofilm (PL- Selection of the study group
biofilm).
Historically, various microscopy techniques have been The study group was composed of 15 systemically healthy
used to visualize the PL-biofilm microstructure, including adult volunteers between 20 and 45 years old and who pre-
optic microscopy, transmission microscopy, and scanning sented a good oral health status: a minimum of 24 permanent
electron microscopy [17]. Samples are distorted using these teeth with no evidence of gingivitis or periodontitis (commu-
techniques, making correct analysis very difficult, especially nity periodontal index score = 0) [28] and an absence of
in fluid-filled structures [18]. These problems have been elim- untreated caries at the beginning of the study. The following
inated or at least reduced with confocal laser scanning exclusion criteria were applied: smoker or former smoker,
microcopy (CLSM). Its main advantage is permitting PL- presence of dental prostheses or orthodontic devices, antibi-
biofilm analysis without altering its delicate structure [19]; otic treatment or routine use of oral antiseptics in the previous
in addition, this technique facilitates observation in real 3 months, and presence of any systemic disease that could
time. It also permits acquiring very thin optical sections alter the production or composition of the saliva. Professional
(0.52 m) and examining the XZ and XY relationships tooth cleaning was performed on all volunteers before starting
existing between the bacteria and its environment, significant- the study.
ly improving, at the same time, the lateral resolution [20]. This project was approved (number 2012/394) by the
In contrast to traditional microbial quantification methods, Clinical Research Ethics Committee of Galicia. Written in-
systems based on fluorescence have gained increasing impor- formed consent was obtained from all participants in the study.
tance since they are accepted as a simple, precise, reproduc-
ible, and highly sensitive procedure for quantifying adhered Production of the Intraoral Device of Overlaid Disk-holding
microorganisms [21]. Furthermore, although there is not a Splints (IDODS)
standard classification of the different bacterial states of vital-
ity, the staining capacity of the dyes present in the live/dead After considering a number of previously described in situ
assays seems to match with the physiological state of the models [8, 19, 24, 25], an individualized splint of the lower
bacteria, though there are still intermediate colors with un- arch was created for each volunteer, which was able to hold
known interpretation [22]. six glass disks (6 mm in diameter, 1 mm thickness) and
As a result, fluorescence staining has been incorporated to polished at 4,000 grit. The characteristics of this splint have
analyze biofilm structure and vitality using a wide variety of been previously described by other authors [29], although a
dye combinations having been used in PL-biofilm studies in new variety of splint has now been introduced; the old one
situ [8, 2326] due to their ability to stain live and dead was a complete individual inferior arch splint, which has now
bacteria in a selective manner. The combination of SYTO 9 been broken in two, going from the last molar to the
and propidium iodide has been posed as a reliable alternative homolateral canine (Figs. 1 and 2). These splints are formed
[27] to traditional blend of fluorescein diacetate with ethidium with two sheets, an internal 1-mm soft vinyl sheet where the
bromide, mainly because of the destructive properties of this disks are attached and an external 1-mm rigid one made of
combination and the toxicity and instability of ethidium bro- polyethylene terephtalate that is fenestrated.
mide [27]. This staining method allows for a visual demon- The splints with the glass disks were worn by the volun-
stration of bactericidal activity as well as its quantification teers for 48 h to favor growth of the PL-biofilm, withdrawing
using computerized image analysis [12]. it from the oral cavity only during meals (it was stored in an
Clin Oral Invest (2015) 19:97107 99

Processing of the samples of PL-biofilm

The characteristics of LIVE/DEAD BacLight fluorescence


solution (Molecular Probes, Leiden, The Netherlands), as well
as its preparation, have been described by authors in a previ-
ous study [30]. The glass disks were withdrawn from the
splint and were immediately submerged in 100 L of fluores-
cence solution and kept in a dark chamber at room tempera-
ture for 15 min. Microscopic observation was performed by a
Fig. 1 Design of the intraoral device of overlaid disk-holding splints single investigator, who was unaware of the study design,
(partial model). 1 Inner splint in contact with the lower dental arch with using a Leica TCS SP2 laser scanning spectral confocal mi-
holes on the disk area. 2 Glass disk. 3 Outer splint with holes on the disk
area croscope (Leica Microsystems Heidelberg GmbH,
Mannheim, Germany) with an HCX APOL 63x/0.9 water
immersion lens.
opaque container in humid conditions) and to perform oral Four selected fields or XYZ series in the central part of
hygiene procedures, using only mechanical removal of bacte- each disk were evaluated. These fields were considered as
rial plaque with water without the use of any toothpaste or representative of the whole sample after the observers general
mouthwash. examination. Fluorescence emission was determined in series
of XY images in which each image corresponded to each of
the Z positions (depth). The optical sections were scanned in
Application of the essential oil to PL-biofilm 1-m sections from the surface of the biofilm to its base,
measuring the maximum thickness of the field and subse-
After 48 h, the glass disks were withdrawn one by one from quently the mean thickness of the biofilm of the corresponding
the splint from each volunteer (from right to left in a distal sample. The maximum thickness of biofilm field was defined
mesial direction) at baseline, 30 s, and 1, 3, 5, and 7 h after as the distance between the substrate and the peaks of the
performing the following mouthwashes under supervision: highest cell clusters [31]. The maximum biofilm thickness of
each field was divided into three zones or equivalent layers:
1. A single, 30-s mouthwash with 20 mL of sterile water outer layer (layer 1), middle layer (layer 2), and inner layer
(negative control) (M-WATER) or (layer 3).
2. A single, 30-s mouthwash with 10 mL of 0.2 % chlorhex- Quantification of bacterial vitality in the series of XY
idine (Oraldine Perio, Johnson and Johnson, Madrid, images was determined using cytofluorographic analysis
Spain) (positive control) (M-0.2 % CHX) or (Leica confocal software). In this analysis, the images of each
3. A single, 30-s mouthwash with 20 mL of essential oils in fluorochrome were defined as channels (SYTO 9 occupies
a hydroalcoholic solution (Listerine Menthol, Listerine, the green channel and PI the red channel), obtaining values for
Johnson & Johnson, Madrid, Spain) (M-EO) the area (m2) occupied by each channel, the total area
occupied by the biofilm, and the corresponding percentage
On the day of the experiment, the volunteers were not of vitality. Determination of the mean percentage of bacterial
allowed to eat or drink during the course of the tests. vitality in each field required sections with a minimum area of
Collection of the different PL-biofilm samples started at biofilm of 250 m2, and the mean percentage of bacterial
11:50 AM (baseline sample) and finished at 7:00 PM (the final vitality of the biofilm was calculated for the corresponding
sample was obtained 7 h after performing the mouthwash). sample and for each biofilm layer.
Using an Internet-based balanced randomization system
(Dallal GE www.randomization.com) indicating the Statistical analysis
mouthwash each subject would use first, second, and third,
all volunteers performed the three mouthwashes, with a rest The results were analyzed using the PASW Statistics Base
period of 2 weeks between each test. 18 package for Windows (IBM, Madrid, Spain). Data on

Fig. 2 Clinical images of the


partial splint model
100 Clin Oral Invest (2015) 19:97107

thickness and bacterial vitality in PL-biofilms are expressed as all the biofilm samples taken after M-EO (p<0.05 in all
mean and standard deviation of the mean. All values from the comparisons). In comparison with M-0.2 % CHX, the preva-
quantitative variables analyzed (biofilm thickness and bac- lence of live bacteria was significantly lower in the middle and
terial vitality percentage) presented a normal distribution, inner layers from 1 h after mouthwash use to 7 h later
which was determined using the KolmogorovSmirnov test. (Table 2).
One-way ANOVA with repeated measures was used for intra-
mouthwash comparisons using all the PL-biofilm samples.
Two-way ANOVA with repeated measures was used for Discussion
intra-mouthwash (differentiating between the three biofilm
layers) and inter-mouthwash comparisons using all the PL- Methodological approach
biofilm samples. Three-way ANOVA with repeated measures
was used for inter-mouthwash (differentiating between the There has been marked inter-individual variability detected
three biofilm layers) comparisons using all the PL-biofilm regarding the characteristics of PL-biofilm [7, 18, 19, 26, 32].
samples. Pairwise comparisons (with Bonferroni adjustment) In the present study, a sample group of 15 individuals was
were used for the analysis of intra- and inter-mouthwash selected. This sample group is bigger than in similar studies in
results (including differentiating between the three biofilm which the number of volunteers ranged from 3 to 10 [15, 16,
layers). Statistical significance was taken as a p value less 19, 33, 34]. With regard to the type of removable appliance
than 0.05. used to collect the supragingival dental plaque, devices such
as the Leeds in situ device [9, 18, 35, 36], bilateral mandibular
stents [32, 37, 38], and different types of individualized acryl-
Results ic splints [7, 8, 10, 19, 23, 25] have been previously described.
In the present series, two individualized splints formed from
Influence of 0.2 %-CHX and M-EO on PL-biofilm thickness two sheets were designed for each volunteer. The splint was
composed of an internal vinyl sheet to which three disks were
The mean PL-biofilm thickness at baseline was 22.1 m attached, with an external polyethylene terephtalate sheet that
(range 1228 m). Significant differences were not found was fenestrated to permit contact between the vestibular sur-
over time after M-EO with regard to basal thickness. On the face of the disks and the saliva whilst protecting the surface
other hand, after M-0.2 % CHX, lower PL-biofilm thickness from the action of the cheeks and tongue. The disks were
values were obtained in comparison to both the basal thick- positioned on each hemiarch and inserted towards the inter-
ness and the M-EO thickness (Table 1). dental area between two adjacent teeth in order to imitate an
approximate PL-biofilm, which is only minimally influenced
Influence of 0.2 %-CHX and M-EO on PL-biofilm bacterial by the shear forces of the oral soft tissues. This particular
vitality design ensured that the biofilm was not touched or disturbed
during removal or repositioning of the appliance [29]. In
The basal vitality in PL-biofilm was 73.6 % (4494 %). The contrast to previous designs [7, 8, 19], where the splints
M-WATER mouthwash did not have any significant effect on referred to a complete model, the partial model of IDODS
PL-biofilm vitality compared to the basal level. The results represents a new approach in the way of making a better in situ
after M-0.2 % CHX and M-EO showed significant differences biofilm model. This redesign was more comfortable for the
compared to their respective basal levels from 30 s after participants when talking and wearing the splints due to the
mouthwash use to 7 h later (Fig. 3). In comparison with the fact that their incisors were not covered. At the same time, the
values obtained, 30 s after M-0.2 % CHX and M-EO, a extraction of the disks was easier because it was not necessary
significant recovery of the bacterial population was observed to remove the whole inferior arch splint but only the hemiarch
in the later PL-biofilm samples (after 3 and 5 h, respectively). corresponding to the analysis, keeping the other undisturbed.
Comparing M-0.2 % CHX and M-EO, M-EO presented lower A number of solid substrates of different characteristics
percentages of bacterial vitality up to 7 h after application, have been used in published studies on PL-biofilm, including
obtaining significant differences from 1 to 5 h post- human enamel [18, 24, 25, 38], bovine enamel [10, 23, 34],
mouthwash (Fig. 4). bovine dentine [26, 34], hydroxyapatite [15], polished glass
Differentiating between the three biofilm layers, the prev- [7, 8, 19, 24], and titanium [16]. Although the roughness of
alence of live bacteria under basal conditions was higher in the the surface of the substrate and its free energy are considered
outer layers with respect to deeper layers, reaching statistical to be important factors for the in vivo growth of PL-biofilm
significance in the majority of comparisons (Table 2). [7], Netuschil et al. [24] found no major differences in the
In comparison with M-WATER, the prevalence of live thickness of 2-day PL-biofilm using enamel or glass disks; on
bacteria was significantly lower in the three biofilm layers in the other hand, due to the known autofluorescence of enamel,
Table 1 Measurement of PL-Biofilm thickness, as well as intra-mouthwash and inter-mouthwash comparisons, before the different mouthwashes (baseline) and after (30 seconds, 1 hour, 3 hours, 5 hours,
and 7 hours)

PL-Biofilm THICKNESS MeanStandard Deviation (m)


BASAL 30 SEC 1H 3H 5H 7H
M-WATER 19.35.4 18.02.6 22.35.4 21.05.1 23.94.8 23.93.9
M-0.2 % CHX 23.48.3 15.81.9 13.52.5 15.42.9 17.53.9 15.62.3
M-EO 23.64.7 20.93.6 20.24.3 21.04.5 18.84.4 21.62.8
INTRA-MOUTHWASH ANALYSIS
Clin Oral Invest (2015) 19:97107

BASAL vs. 30 SEG BASAL vs. 1 H 30 SEC vs. 1 H BASAL vs. 3 H 30 SEC vs. 3 H BASAL vs. 5 H 30 SEC vs. 5 H BASAL vs. 7 H 30 SEC vs. 7 H
M-WATER p<0.05 p<0.05 p<0.05
M-0.2 % CHX p<0.05 p<0.05 p<0.05
M-EO
INTER-MOUTHWASH ANALYSIS
BASAL 30 SEC 1H 3H 5H 7H
M-WATER vs. M-EO p<0.05
M-0.2 % CHX vs. M-EO p<0.001 p<0.001 p<0.05 p<0.001

Not a statistically significant difference.


M-WATER=a single, 30-second mouthwash with 20 mL of sterile water; M-0.2 % CHX=a single, 30-second mouthwash with 10 mL of 0.2 % chlorhexidine; M-EO=a single, 30-second mouthwash with
20 mL of essential oil solution.
BASAL=Biofilm sample collected under basal conditions; 30 SEC=Biofilm sample collected 30 seconds after the application of the different mouthwashes; 1 H=Biofilm sample collected 1 hour after the
application of the different mouthwashes; 3 H=Biofilm sample collected 3 hours after the application of the different mouthwashes; 5 H=Biofilm sample collected 5 hours after the application of the
different mouthwashes; 7 H=Biofilm sample collected 7 hours after the application of the different mouthwashes.
101
102 Clin Oral Invest (2015) 19:97107

Fig. 3 Representative images of the PL-biofilm bacterial vitality under basal M-0.2 % CHX, and M-EO, respectively. df Images taken 30 s after M-
conditions and at 30 s and 7 h after the application of different mouthwashes. WATER, M-0.2 % CHX, and M-EO, respectively. gh Images taken 7 h
ac Basal samples collected before different mouthwashes with M-WATER, after M-WATER, M-0.2 % CHX, and M-EO, respectively

using glass is recommended to avoid any optical disturbance, agents activity, although they continued to ask the question
mainly in the deepest layers of the biofilm [24]. On the basis how dead is dead? due to several stages of vitality which
of these findings, in the present series, glass disks were used have been discussed and described in the literature (viable and
for in vivo growth of the 2-day PL-biofilm. culturable, viable but non-culturable, dormant, non-viable and
In a recent paper, Tawakoli et al. [27] stated that the pre-lytic, and avital dead bacteria). The exact differentiation of
BacLight system was a reliable alternative when assessing these stages is still one of the greatest challenges in modern
bacterial vitality in a 120-min-old natural dental biofilm, in microbiology [40].
which there are already several types of bacteria present. The In addition, Tawakoli et al. [27] said that it was not possible
LIVE/DEAD BacLight fluorescence assay stains the bac- to compare properly the vitality assessed with fluorescence
teria in red or green depending on the permeability of their staining solutions with traditional plaque cultures because of
membrane; given that the tested antiseptics act mostly at this the widely known limitations of this later method (among
cellular element, this vital staining method is suitable for this others, only 50 % of oral bacteria are culturable), which
type of study. emphasizes the necessity of using vitality assays. On the other
Furthermore, Hannig et al. [39] considered that live/dead hand, the authors recognize the convenience of contrasting
staining methods were reliable when analyzing antimicrobial and complementing the data obtained with BacLight
Clin Oral Invest (2015) 19:97107 103

Fig. 4 PL-biofilm bacterial vitality percentage under basal conditions and at 30 s and 1, 3, 5, and 7 h after a single water mouthwash (M-WATER), 0.2 %
chlorhexidine mouthwash (M-0.2% CHX), or essential oil (M-EO)

fluorescence solution with other molecular or bacteriological and embedded in dead layers, which may be responsible for
techniques such as the plate efficiency. further plaque growth [24].
In some series, large inter-individual differences were
M-EO: immediate effect, substantivity, and influence found among the subjects in their PL-biofilm vitality distribu-
on PL-biofilm thickness tion [26], so no general pattern for the bacterial vitality distri-
bution could be described [26, 41]; in the present study, the
Studies which have analyzed in situ PL-biofilm have empha- PL-biofilm vitality ranged from 44 to 90 %. However, it has
sized the great variation detected in PL-biofilm thickness been suggested that a relatively constant ecological environ-
between individuals [7, 25, 33]; this was also observed in ment exists in each volunteer, which obviously leads to a
the present study (mean value of PL-biofilm thickness after microbial identity pattern [19]. In this sense, Arweiler et al.
2 days was 22.10 m, ranging from 12 to 28 m), indicating [19] detected great variation in the bacterial vitality values in a
that, in agreement with previous studies, the height of the oral 2-day PL-biofilm for the different biofilm layers, identifying
biofilms formed depended on the plaque-forming rate of the three vitality patterns. In this study, despite the high degree of
individual donors [25]. Our mean value of PL-biofilm was variability detected in the bacterial vitality distribution, a
consistent with that obtained by Dong et al. [15] under similar vitality pattern could be identified, which was based on a
conditions, which was 27.55 m. low vitality percentage observed in the layers nearest to the
In the present series, the bacterial vitality of PL-biofilm was substrate, increasing in higher layers. This finding confirms
approximately 73 %. These results are consistent with previ- the importance of the dead cellular material in the initial states
ous studies which reported mean bacterial vitality of PL- of PL-biofilm development. This will particularly help its
biofilm between 60 and 77 % over 2- and 3-day periods [8, growth, and this material will protect it from antibacterial
19, 41]. Consequently, vital micro-organisms were located on agents in the oral cavity [24, 25].
104 Clin Oral Invest (2015) 19:97107

Table 2 Mean percentages of bacterial vitality in PL-biofilm under basal between the three biofilm layers, as well as intra-mouthwash and inter-
conditions and in the samples collected at 30 s and 1, 3, 5, and 7 h after a mouthwash comparisons
single mouthwash of sterile water and 0.2 % chlorhexidine differentiating

BASAL 30 SEC 1H 3H 5H 7H

PL-biofilm bacterial vitality divided in layers, meanstandard deviation (%)


M-WATER
Layer 1 85.46.6 85.313.1 88.39.6 90.88.9 89.08.5 90.95.9
Layer 2 79.87.3 73.115.1 78.416.6 84.410.3 81.312.8 85.27.1
Layer 3 66.827.3 45.833.4 49.429.8 56.731.5 55.424.9 62.024.0
M-0.2 %CHX
Layer 1 80.06.2 5.216.2 15.115.4 35.415.5 21.719.8 27.022.6
Layer 2 82.27.8 5.066.4 16.515.6 36.716.4 24.820.6 28.720.8
Layer 3 71.817.4 5.05.0 15.210.2 35.214.8 27.513.5 40.020.4
M-EO
Layer 1 78.710.7 1.71.6 5.04.3 13.714.0 15.813.2 37.519.1
Layer 2 69.016.5 1.11.2 2.93.3 6.010.2 5.45.3 12.59.9
Layer 3 49.136.0 0.70.6 4.16.8 5.08.2 3.67.0 3.22.9
Intra-mouthwash analysis
M-WATER
Layer 1 vs. Layer 2 p<0.05 p<0.001 p<0.001 p<0.001 p<0.001 p<0.001
Layer 1 vs. Layer 3 p<0.001 p<0.001 p<0.001 p<0.001 p<0.001
Layer 2 vs. Layer 3 p<0.001 p<0.001 p<0.001 p<0.001 p<0.001
M-CHX 0.2 %
Layer 1 vs. Layer 2
Layer 1 vs. Layer 3 p<0.05
Layer 2 vs. Layer 3 p<0.05 p<0.05
M-EO
Layer 1 vs. Layer 2 p<0.05 p<0.05 p<0.05 p<0.001 p<0.05 p<0.001
Layer 1 vs. Layer 3 p<0.05 p<0.05 p<0.001
Layer 2 vs. Layer 3 p<0.05 p<0.05
Inter-mouthwash analysis
M-WATER vs. M-EO
Layer 1 vs. Layer 1 p<0.001 p<0.001 p<0.001 p<0.001 p<0.001
Layer 2 vs. Layer 2 p<0.001 p<0.001 p<0.001 p<0.001 p<0.001
Layer 3 vs. Layer 3 p<0.001 p<0.001 p<0.001 p<0.001 p<0.001
M-CHX 0.2 % vs. M-EO
Layer 1 vs. Layer 1 p<0.05
Layer 2 vs. Layer 2 p<0.05 p<0.05 p<0.001 p<0.05
Layer 3 vs. Layer 3 p<0.05 p<0.05 p<0.001 p<0.001 p<0.001

not a statistically significant difference, M-WATER a single, 30-s mouthwash with 20 mL of sterile water, M-0.2% CHX a single, 30-s mouthwash with
10 mL of 0.2 % chlorhexidine, M-EO a single, 30-s mouthwash with 20 mL of essential oil solution, BASAL biofilm sample collected under basal
conditions, 30 SEC biofilm sample collected 30 s after the application of the different mouthwashes, 1 H biofilm sample collected 1 h after the application
of the different mouthwashes, 3 H biofilm sample collected 3 h after the application of the different mouthwashes, 5 H biofilm sample collected 5 h after
the application of the different mouthwashes, 7 H biofilm sample collected 7 h after the application of the different mouthwashes, Layer 1 outer layer,
Layer 2 middle layer, Layer 3 inner layer

Although some papers have been published on EO antimi- since our research group has deeply analyzed and discussed
crobial activity on biofilm in vitro [36, 42, 43], studies on the 0.2 % CHX antimicrobial activity on in situ PL-biofilm in
in situ effects of EO on PL-biofilm applying CLSM and previous publications.
bacterial vitality techniques are very scarce. In this study, the Up to now, there have been few papers which have studied
results with M-0.2 % CHX were taken as a positive control the in situ antimicrobial effect on biofilm after a single
Clin Oral Invest (2015) 19:97107 105

application of EO. In two cases, the treatment was practiced mouthwash was more effective at maintaining low values of
ex vivo, which means that no mouthwash was done [15, 16]. bacterial vitality. These findings are not described in the
In the other three [1214], the studied plaque was completely available literature due to the lack of studies measuring the
disturbed by the recollection method (paper points or cu- substantivity of EO in oral biofilm in comparison with CHX.
rettes). Furthermore, only one paper has been published in Another interesting aspect of this study is the higher pen-
which the authors compared EO and CHX antimicrobial etration capacity into the PL-biofilm of EO compared to 0.2 %
activity after a single application (application ex vivo), and CHX. Statistical differences were found in bacterial vitality
they only measured the immediate antimicrobial effect [16]. reduction in layer 3 (the deepest one) between both antisep-
Consequently, the present results have been compared with tics, starting from the immediate and 1-h samples, but more so
those obtained in other studies that applied different when time passed and until 7 h post-mouthwash. This indi-
methodologies. cates that the penetration capacity of a single M-EO applica-
tion is greater than that shown by a single application of
Immediate effect and substantivity M-0.2 % CHX. As Pan et al. [12] previously described, these
results confirm the ability of the essential oil mouthwash to
With a similar basal vitality between the three mouthwashes rapidly penetrate plaque and exert its bactericidal activity in
(mean of 73.6 %), a clear and immediate post-mouthwash situ. Apart from that, the M-EO maintains its antimicrobial
effect was detected after EO application. This immediate activity in the deepest layers (closest to the theoretical tooth
activity was very high if compared to that obtained in a similar surface) for a longer time.
study reported by Gosau et al. [16] since, in the previous There have been other in vivo studies demonstrating the
study, bacterial vitality after M-EO was around 20 %, while efficacy of a single mouthwash application in other oral eco-
in the present series, the vitality was 1 % 30 s after M-EO. The systems, such as the saliva. It has been shown that a single M-
methodological differences should be noted; in the previous EO or M-0.2 % CHX application can significantly reduce the
case, ex vivo disk immersion was performed, while in the levels of recoverable salivary bacteria compared to negative
present series, the volunteers themselves used the mouthwash, control mouthwashes for periods of 57 h [31, 44]. Both
providing in vivo antiseptic application. This is evidently a studies showed higher bacterial vitality than in PL-biofilm,
more reliable approximation of the clinical situation. in comparison with the present series, so a longer antiseptic
Therefore, it was found that moving the essential oil solution (both EO and 0.2 % CHX) substantivity period was detected
around the mouth and the force imposed by the cheeks when in the PL-biofilm.
projecting it onto the PL-biofilm is probably of prime impor-
tance for reducing bacterial biofilm vitality. In the same way, Thickness reduction
this could be also related to improved deep penetration com-
pared to when the mouthwash was not actively applied. As As the results of the present series show, a single M-EO
expected, the negative control (M-WATER) had no antibacte- application is not effective in reducing PL-biofilm thickness.
rial activity compared to M-EO. In comparison with the These results are consistent with those previously described
positive control (M-0.2 % CHX), a single application of M- by Dong et al. [15], who did not find significant differences in
EO was more effective, consistent with previous results ob- biofilm thickness with regard to the basal sample after apply-
tained by other groups [3, 16]. ing M-EO. These results are also consistent with an in vitro
In the present series, EO antimicrobial activity was detect- study conducted by Sliepen et al. [3] who concluded that EO
able until 7 h after mouthwash application, when the reduction caused nearly no changes in biofilm structure, thickness, and
in vitality was still 61 %. In this study, it was also appreciated surface coverage. With respect to M-0.2 % CHX, statistically
that a single M-EO application was effective for maintaining significant differences were found compared to M-EO at 30 s
low bacterial vitality levels in PL-biofilm. As shown in Fig. 4, and 1, 3, and 7 h after mouthwash use, which could suggest a
there were no statistical differences in bacterial vitality until possible antiplaque effect of 0.2 % CHX after a single anti-
5 h compared to the mouthwash sample after 30 s, which septic application.
indicates powerful antibacterial activity at high levels until Moreover, the group of Charles [13] concluded that the
that moment. Up to now, there have been two studies on the clinical effectiveness of a single M-EO application against
substantivity of essential oils in oral biofilm. The first found plaque and gingivitis may be attributable to its bactericidal
21.3 % vitality 30 min post-mouthwash [12], while the other, effect and penetration into the dental plaque. Subsequently,
conducted by Fine et al. [14], showed a vitality reduction of other authors demonstrated that performing daily mouth-
88 % after 12 h. Both studies used a disturbed dental plaque washes with an EO solution has a considerable antiplaque
in vivo model, so these results are not fully comparable. effect [45, 46]. However, two recent literature reviews [47,
Compared with the positive control, there were significant 48] concluded that daily essential oil mouthwash use has a
differences from 1 to 5 h post-mouthwash, and the EO lower antiplaque effect than using 0.2 % CHX, although the
106 Clin Oral Invest (2015) 19:97107

gingival inflammation levels were quite similar in the long 9. Watson PS, Pontefract HA, Devine DA, Shore RC, Nattress BR,
Kirkham J, Robinson C (2005) Penetration of fluoride into natural
and short term. Based on these findings, it would be very
plaque biofilms. J Dent Res 84(5):451455
interesting to analyze the antiplaque effect associated with 10. Al-Ahmad A, Wunder A, Auschill TM, Follo M, Braun G, Hellwig
continuous EO use in a 4-day in situ undisturbed PL- E, Arweiler NB (2007) The in vivo dynamics of Streptococcus spp.,
biofilm model as the next step in this research. Actinomyces naeslundii, Fusobacterium nucleatum and Veillonella
spp. in dental plaque biofilm as analysed by five-colour multiplex
fluorescence in situ hybridization. J Med Microbiol 56(Pt 5):681
687. doi:10.1099/jmm.0.47094-0
11. Hannig C, Hannig M (2009) The oral cavitya key system to
Conclusion
understand substratum-dependent bioadhesion on solid surfaces in
man. Clin Oral Investig 13(2):123139. doi:10.1007/s00784-008-
A single application of essential oil mouthwash presents high 0243-3
antibacterial immediate activity and penetration capacity in 12. Pan P, Barnett ML, Coelho J, Brogdon C, Finnegan MB (2000)
Determination of the in situ bactericidal activity of an essential oil
situ and substantivity which lasts, at least, for 7 h after its
mouthrinse using a vital stain method. J Clin Periodontol 27(4):256
application over de novo biofilm. These results are better than 261. doi:10.1034/j.1600-051x.2000.027004256.x
those observed with 0.2 % chlorhexidine under the same 13. Charles CH, Pan PC, Sturdivant L, Vincent JW (2000) In vivo
conditions. antimicrobial activity of an essential oil-containing mouthrinse on
interproximal plaque bacteria. J Clin Dent 11(4):9497
14. Fine DH, Furgang D, Sinatra K, Charles C, McGuire A, Kumar LD
Acknowledgments This work was supported by project PI11/01383 (2005) In vivo antimicrobial effectiveness of an essential oil-
from Carlos III Institute of Health (General Division of Evaluation and containing mouth rinse 12 h after a single use and 14 days use. J
Research Promotion, Madrid, Spain), which is integrated in National Plan Clin Periodontol 32(4):335340. doi:10.1111/j.1600-051x.2005.
of Research, Development and Innovation (PN I + D + I 2008-2011). This 00674.x
project was co-financed by European Regional Development Fund 15. Dong WL, Zhou YH, Li CZ, Liu H, Shang SH, Pan BQ (2010)
(ERDF 20072013). The funders had no role in the study design, data Establishment and application of an intact natural model of human
collection and analysis, decision to publish, or preparation of the dental plaque biofilm. Shanghai Kou Qiang Yi Xue 19(2):196201
manuscript. 16. Gosau M, Hahnel S, Schwarz F, Gerlach T, Reichert TE, Burgers R
(2010) Effect of six different peri-implantitis disinfection methods on
Conflict of interest The authors declare that they have no conflict of in vivo human oral biofilm. Clin Oral Implants Res 21(8):866872.
interest. doi:10.1111/j.1600-0501.2009.01908.x
17. Hannig C, Hannig M, Rehmer O, Braun G, Hellwig E, Al-Ahmad A
(2007) Fluorescence microscopic visualization and quantification of
initial bacterial colonization on enamel in situ. Arch Oral Biol 52(11):
References 10481056
18. Wood SR, Kirkham J, Marsh PD, Shore RC, Nattress B, Robinson C
(2000) Architecture of intact natural human plaque biofilms studied
1. Vlachojannis C, Winsauer H, Chrubasik S (2012) Effectiveness and by confocal laser scanning microscopy. J Dent Res 79(1):2127. doi:
safety of a mouthwash containing essential oil ingredients. Phytother 10.1177/00220345000790010201
Res 27(5):685691. doi:10.1002/ptr.4762 19. Arweiler NB, Hellwig E, Sculean A, Hein N, Auschill TM (2004)
2. FDA (2003) Oral health care drug products for over-the-counter Individual vitality pattern of in situ dental biofilms at different loca-
human use; antigingivitis/ antiplaque drug products; establishment tions in the oral cavity. Caries Res 38(5):442447. doi:10.1159/
of a monograph; proposed rules part III. Vol 21 CFR 000079625
3. Sliepen I, Van Essche M, Quirynen M, Teughels W (2010) Effect of 20. Wright SJ, Wright DJ (2002) Introduction to confocal microscopy.
mouthrinses on Aggregatibacter actinomycetemcomitans biofilms in Methods Cell Biol 70:185
a hydrodynamic model. Clin Oral Investig 14(3):241250. doi:10. 21. Hahnel S, Rosentritt M, Burgers R, Handel G (2008) Surface prop-
1007/s00784-009-0286-0 erties and in vitro Streptococcus mutans adhesion to dental resin
4. Fine DH, Furgang D, Barnett ML (2001) Comparative antimicrobial polymers. J Mater Sci Mater Med 19(7):26192627. doi:10.1007/
activities of antiseptic mouthrinses against isogenic planktonic and s10856-007-3352-7
biofilm forms of Actinobacillus actinomycetemcomitans. J Clin 22. Berney M, Hammes F, Bosshard F, Weilenmann HU, Egli T (2007)
Periodontol 28(7):697700. doi:10.1034/j.1600-051x.2001. Assessment and interpretation of bacterial viability by using the
028007697.x LIVE/DEAD BacLight Kit in combination with flow cytometry.
5. Filoche SK, Coleman MJ, Angker L, Sissons CH (2007) A fluores- Appl Environ Microbiol 73(10):32833290. doi:10.1128/AEM.
cence assay to determine the viable biomass of microcosm dental 02750-06
plaque biofilms. J Microbiol Methods 69(3):489496 23. Arweiler NB, Lenz R, Sculean A, Al-Ahmad A, Hellwig E, Auschill
6. Pan PC, Harper S, Ricci-Nittel D, Lux R, Shi W (2010) In-vitro TM (2008) Effect of food preservatives on in situ biofilm formation.
evidence for efficacy of antimicrobial mouthrinses. J Dent 38(Suppl Clin Oral Investig 12(3):203208. doi:10.1007/s00784-008-0188-6
1):S16S20. doi:10.1016/S0300-5712(10)70006-3 24. Netuschil L, Reich E, Unteregger G, Sculean A, Brecx M (1998) A
7. Auschill TM, Hellwig E, Sculean A, Hein N, Arweiler NB (2004) pilot study of confocal laser scanning microscopy for the assessment
Impact of the intraoral location on the rate of biofilm growth. Clin of undisturbed dental plaque vitality and topography. Arch Oral Biol
Oral Investig 8(2):97101. doi:10.1007/s00784-004-0255-6 43(4):277285. doi:10.1016/S0003-9969(97)00121-0
8. Auschill TM, Hein N, Hellwig E, Follo M, Sculean A, Arweiler NB 25. Auschill TM, Arweiler NB, Netuschil L, Brecx M, Reich E, Sculean
(2005) Effect of two antimicrobial agents on early in situ biofilm A (2001) Spatial distribution of vital and dead microorganisms in
formation. J Clin Periodontol 32(2):147152. doi:10.1111/j.1600- dental biofilms. Arch Oral Biol 46(5):471476. doi:10.1016/S0003-
051X.2005.00650.x 9969(00)00136-9
Clin Oral Invest (2015) 19:97107 107

26. Zaura-Arite E, van Marle J, ten Cate JM (2001) Confocal microscopy 38. Chalmers NI, Palmer RJ Jr, Du-Thumm L, Sullivan R, Shi W,
study of undisturbed and chlorhexidine-treated dental biofilm. J Dent Kolenbrander PE (2007) Use of quantum dot luminescent probes to
Res 80:14361440. doi:10.1177/00220345010800051001 achieve single-cell resolution of human oral bacteria in biofilms. Appl
27. Tawakoli PN, Al-Ahmad A, Hoth-Hannig W, Hannig M, Hannig C Environ Microbiol 73(2):630636. doi:10.1128/AEM.02164-06
(2013) Comparison of different live/dead stainings for detec- 39. Hannig C, Follo M, Hellwig E, Al-Ahmad A (2010) Visualization of
tion and quantification of adherent microorganisms in the adherent micro-organisms using different techniques. J Med
initial oral biofilm. Clin Oral Investig 17(3):841850. doi:10. Microbiol 59(Pt 1):17. doi:10.1099/jmm.0.015420-0
1007/s00784-012-0792-3 40. Decker EM (2001) The ability of direct fluorescence-based, two-
28. WHO (1997) Oral health surveys, basic methods, 4th edn. WHO, colour assays to detect different physiological states of oral strepto-
Geneva cocci. Lett Appl Microbiol 33(3):188192. doi:10.1046/j.1472-765x.
29. Toms I HB, Diz P, Donos N (2010) In vivo oral biofilm analysis by 2001.00971.x
confocal laser scanning microscopy: methodological approaches. In: 41. von Ohle C, Gieseke A, Nistico L, Decker EM, DeBeer D, Stoodley
A M-V (ed) Microscopy. Science, technology, applications and edu- P (2010) Real-time microsensor measurement of local metabolic
cation. Formatex, Badajoz (Spain), pp 597-606 activities in ex vivo dental biofilms exposed to sucrose and treated
30. Toms I, Garca-Caballero L, Cousido MC, Limeres J, lvarez M, with chlorhexidine. Appl Env Microbiol 76(7):23262334. doi:10.
Diz Dios P (2009) Evaluation of chlorhexidine substantivity on 1128/AEM.02090-09
salivary flora by epifluorescence microscopy. Oral Dis 15(6):428 42. Pan PH, Finnegan MB, Sturdivant L, Barnett ML (1999)
433. doi:10.1111/j.1601-0825.2009.01570.x Comparative antimicrobial activity of an essential oil and an amine
31. Roberts SKBC, Brading M, Lappin-Scott H, Stoodley P (1999) fluoride/stannous fluoride mouthrinse in vitro. J Clin Periodontol
Biofilm formation and structure; whats new? In: Newman HNWM 26(7):474476
(ed) Dental plaque revisitedoral biofilms in health and disease. 43. Fine DH, Furgang D, Barnett ML, Drew C, Steinberg L, Charles CH,
BioLine, Cardiff, pp 136 Vincent JW (2000) Effect of an essential oil-containing antiseptic
32. Diaz PI, Chalmers NI, Rickard AH, Kong C, Milburn CL, Palmer RJ mouthrinse on plaque and salivary Streptococcus mutans levels. J
Jr, Kolenbrander PE (2006) Molecular characterization of subject- Clin Periodontol 27(3):157161. doi:10.1034/j.1600-051X.1999.
specific oral microflora during initial colonization of enamel. Appl 260710.x
Environ Microbiol 72(4):28372848. doi:10.1128/AEM.72.4.2837- 44. Jenkins S, Addy M, Wade W, Newcombe RG (1994) The magnitude
2848.2006 and duration of the effects of some mouthrinse products on salivary
33. Dige I, Nyengaard JR, Kilian M, Nyvad B (2009) Application of bacterial counts. J Clin Periodontol 21(6):397401. doi:10.1111/j.
stereological principles for quantification of bacteria in intact dental 1600-051X.1994.tb00736.x
biofilms. Oral Microbiol Immunol 24(1):6975. doi:10.1111/j.1399- 45. Stoeken JE, Paraskevas S, van der Weijden GA (2007) The long-term
302X.2008.00482.x effect of a mouthrinse containing essential oils on dental plaque and
34. Jung DJ, Al-Ahmad A, Follo M, Spitzmuller B, Hoth-Hannig W, gingivitis: a systematic review. J Periodontol 78(7):12181228. doi:
Hannig M, Hannig C (2010) Visualization of initial bacterial coloni- 10.1902/jop.2007.060269
zation on dentine and enamel in situ. J Microbiol Methods 81(2): 46. Cortelli SC, Cortelli JR, Wu MM, Simmons K, Charles CA (2012)
166174. doi:10.1016/j.mimet.2010.03.002 Comparative antiplaque and antigingivitis efficacy of a multipurpose
35. Wood S, Nattress B, Kirkham J, Shore R, Brookes S, Griffiths J, essential oil-containing mouthrinse and a cetylpyridinium chloride-
Robinson C (1999) An in vitro study of the use of photodynamic containing mouthrinse: a 6-month randomized clinical trial.
therapy for the treatment of natural oral plaque biofilms formed Quintessence Int 43(7):e82e94
in vivo. J Photochem Photobiol B 50(1):17 47. Van Leeuwen MP, Slot DE, Van der Weijden GA (2011) Essential
36. Robinson C, Strafford S, Rees G, Brookes SJ, Kirkham J, Shore RC, oils compared to chlorhexidine with respect to plaque and parameters
Watson PS, Wood S (2006) Plaque biofilms: the effect of chemical of gingival inflammation: a systematic review. J Periodontol 82(2):
environment on natural human plaque biofilm architecture. Arch Oral 174194. doi:10.1902/jop.2010.100266
Biol 51(11):10061014. doi:10.1016/j.archoralbio.2006.04.010 48. Neely AL (2012) Essential oil mouthwash (EOMW) may be equiv-
37. Palmer RJ Jr, Gordon SM, Cisar JO, Kolenbrander PE (2003) alent to chlorhexidine (CHX) for long-term control of gingival in-
Coaggregation-mediated interactions of streptococci and actinomy- flammation but CHX appears to perform better than EOMW in
ces detected in initial human dental plaque. J Bacteriol 185(11): plaque control. J Evid Based Dent Pract 12(3 Suppl):6972. doi:10.
34003409. doi:10.1128/JB.185.11.3400-3409.2003 1016/S1532-3382(12)70017-9
Copyright of Clinical Oral Investigations is the property of Springer Science & Business
Media B.V. and its content may not be copied or emailed to multiple sites or posted to a
listserv without the copyright holder's express written permission. However, users may print,
download, or email articles for individual use.

You might also like