You are on page 1of 10

The Microbiologic Profile Associated with

Peri-Implantitis in Humans: A Systematic Review


Mia Rakic, DDS, PhD1/Maria Gabriella Grusovin, DDS2/Luigi Canullo, DDS, PhD3

Purpose: To qualitatively investigate the microbiologic profile in peri-implantitis by systematically reviewing


the published literature on peri-implant infection. Materials and Methods: Searches of the US National
Institutes of Health free digital archives of the biomedical and life sciences journal literature (PubMed)
and The Cochrane Library of the Cochrane Collaboration (CENTRAL), as well as a hand search of other
literature, were conducted to identify articles potentially relevant for the review. Randomized clinical
trials, prospective cohort studies, longitudinal studies, case-control studies, and cross-sectional studies in
humans reporting microbiologic findings in patients with diagnosed peri-implantitis were considered eligible
for this review. Screening, data extraction, and quality assessment were conducted independently and in
duplicate. Results: Twenty-one articles were eligible for inclusion in this review. Early studies focused on the
identification of target periopathogens, whereas more recent studies used advanced molecular techniques
for comprehensive overview of the peri-implantitis–associated microbiome. In summary, the microbiologic
profile in peri-implantitis (1) is complex and variable, (2) consists of gram-negative anaerobic periopathogens
and opportunistic microorganisms in almost the same ratio, (3) is frequently associated with the Epstein-Barr
virus and nonsaccharolytic anaerobic gram-positive rods, (4) is not so strictly associated with Staphylococcus
aureus, and (5) is different from that of periodontitis. A meta-analysis could not be performed because of
the heterogeneity of the reviewed studies. Conclusion: Although a comparison of the published results was
limited because of the inhomogeneity of the studies, it is clear that the microbiologic profile of peri-implantitis
consists of aggressive and resistant microorganisms and is distinct from that of periodontitis. It seems that
the quantitative characteristics of the microflora cohabitants represent the key determinant of disease,
rather than the qualitative composition, which is very similar in healthy and peri-implantitis states. Int J Oral
Maxillofac Implants 2016;31:359–368. doi: 10.11607/jomi.4150

Keywords: bacteria, microbiota, peri-implantitis, periodontitis

P eri-implantitis is a late complication of oral im-


plants characterized by inflammatory bone loss.1
The central pathologic process in peri-implantitis is
cause of peri-implantitis, and it is considered that all
other factors sooner or later act in conjunction with
infection.2–4 Although peri-implantitis was once con-
inflammatory osteoclastogenesis, which could be sidered a counterpart of periodontitis at implant sites,
triggered by infection, lesions of the peri-implant recent studies have indicated that peri-implant pathol-
attachment, excessive biomechanical stress, and/or ogy seems to be more complex and differs from peri-
biocorrosion.2 However, infection remains the major odontal disease pathology.5 This complexity relates
primarily to the structural specificities of peri-implant
tissues and to the implants themselves. Some studies
1 Assistant Professor, Institute for Biological Research
have suggested that the lack of periodontal ligament
“Sinisa Stankovic,” University of Belgrade, Belgrade, Serbia;
Centre for Osteoarticular and Dental Tissue Engineering and cementum makes the peri-implant tissues more
INSERM U791, Faculty of Dental Surgery, University of susceptible to infection and trauma and leads to fast
Nantes, Nantes, France. progression of disease.6–11 Furthermore, the implant
2 Adjunct Professor, Dental School, Vita-Salute San Raffaele
surface oxide layer (so-called “ceramic-like” layer),
University, Milan, Italy.
3 Private Practice, Rome, Italy.
which is the major factor of titanium implant biocom-
patibility, requires certain physicochemical conditions
Correspondence to: Dr Mia Rakic, Institute for Biological to maintain its stability and, hence, implant biocom-
Research “Sinisa Stankovic,” University of Belgrade, patibility. Related to this, the anaerobic peri-implant
Belgrade, Serbia, Bulevar Despota Stefana 142, infection, which creates an acid microenvironment full
11000 Belgrade, Serbia. Email: mia.rakic@ibiss.bg.ac.rs
of reactive host-derived factors (such as free radicals),
©2016 by Quintessence Publishing Co Inc. is considered a physicochemical threat to the stability

The International Journal of Oral & Maxillofacial Implants 359

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

of the oxide layer that could increase inflammatory Search Strategy


bone loss.4 For these reasons, knowledge of the mi- Searches of the U.S. National Institutes of Health free
crobiologic profile associated with peri-implantitis digital archives of biomedical and life sciences journal
(which would guide infection control and supportive literature (PubMed) and The Cochrane Library of the
periodontal therapy measures) seems to be one of Cochrane Collaboration (CENTRAL), as well as a hand
the factors that is essential for the success of implant search of other literature, were conducted to identify
treatment, particularly in subjects with a history of articles of potential relevance. The search included
periodontitis. Despite this great necessity, the micro- articles accepted for publication up to August 2014.
biologic profile of peri-implantitis has not yet been Previously published review articles on similar topics
clearly defined.13–15 were also analyzed to assess potentially relevant pub-
The objective of this review was to qualitatively lications. The following key words were used for this
estimate the microbiologic profile associated with purpose: periimplantitis OR peri-implantitis OR peri
peri-implantitis in humans through a review of the implantitis OR peri-implant AND infection OR bacteria
literature. OR microorganism OR biofilm OR plaque.

Quality Assessment
MATERIALS AND METHODS Quality assurance was developed according to Khan
et al16 via independent screening by two reviewers,
This systematic review complies with the PRISMA guide- resolution of disagreement by consensus, discarding
lines (Preferred Reporting Items for Systematic Reviews of studies when consensus was not achieved, and data
and Meta-Analyses; www.prisma-statement.org). extraction in duplicate.

Study Protocol and Criteria Data Extraction and Synthesis


The protocol was designed to answer the following Two independent reviewers (MR, LC) analyzed titles
question: “What are the characteristics of the microbi- and abstracts in the first stage of screening. Irrelevant
ologic profile in patients with peri-implantitis?” Includ- articles were discarded. Then, the full texts of the ar-
ed were randomized clinical trials, prospective cohort ticles considered to be potentially relevant for the
studies, case-control studies, and cross-sectional stud- review were read to determine whether the studies
ies in humans reporting microbiologic findings in pa- fulfilled the predetermined inclusion criteria. Any dis-
tients diagnosed with peri-implantitis. Peri-implantitis agreement regarding eligibility of the articles was in-
was defined as the radiographic presence of bone loss dividually resolved between the reviewers. Data were
≥ 2 mm since the time of prosthetic replacement, posi- collated into evidence tables and grouped according
tive bleeding on probing, and probing depth ≥ 5 mm. to the microbiologic analyses that were performed:
Only studies published in English were included. Ex- (1) evaluation of target pathogens or (2) evaluation
cluded were in vitro and animal studies and studies of of the entire microbiome. Furthermore, the extracted
blade implants. data were stratified and expressed in chronologic or-
Furthermore, the following PECO (population, der according to the publication date. Synthesis of
exposure, comparison, outcome) definitions were the data was performed based on the evidence tables
considered. alone, and the data were further interpreted in relation
to the performed study design (cross-sectional, case-
• Population. Studies had to include systemically control, or longitudinal). Meta-analysis was not carried
healthy patients with at least one implant affected out because of the marked heterogeneity of many as-
by peri-implantitis; microbiologic findings from pects of the included studies.
affected sites had to be provided.
• Exposure. Peri-implantitis was the exposure consid-
ered for evaluation. RESULTS
• Comparison. The specific comparisons investigated
were either differences throughout the course The initial search of the literature up to June 2014 yield-
of peri-implantitis or differences between peri- ed 631 potentially suitable papers. Following exclusion
implantitis and peri-mucositis, healthy peri-implant of reviews, animal and in vitro studies, and studies that
tissues, or periodontitis. inappropriately identified peri-implantitis, 21 publica-
• Outcome measures. The primary outcome variable tions remained fully eligible for this review. The κ value
was microbiologic status (total flora, presence of for interreviewer agreement for study inclusion was
certain bacterial pathogens, and percentages and 0.93 for titles and abstracts and 1.00 for full-text arti-
proportions of flora of certain bacterial pathogens). cles, indicating strong agreement. The major findings

360 Volume 31, Number 2, 2016

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

of the reviewed articles were sorted into two tables healthy implants, whereas Capnocytophaga ochracea
according to the type of microbiologic analysis per- was higher in mucositis than in the other groups.23 It was
formed. Table 1 covers the studies that evaluated the also observed that P gingivalis showed the highest levels
target pathogens,17–31 and Table 2 includes studies in peri-implantitis as opposed to the healthy implants, at
that evaluated the entire microbiome.32–37 which red-complex periopathogens were found in very
low levels. Furthermore, a cross-sectional study investi-
Target Pathogens of Peri-implantitis gated microflora in 15 cases of peri-implantitis using cul-
Fifteen studies evaluated target pathogens using cul- tures and established an association among T forsythia,
tures, checkerboard hybridization, polymerase chain Campylobacter sp, and Parvimonas micra with peri-im-
reaction (PCR), or DNA probes. Most studies were plantitis.24 Additionally, a positive correlation between
cross-sectional and case-control studies; only three pain and the presence of P micra, Fusobacterium, and
were longitudinal studies. Eubacterium sp was reported. A more recent cross-sec-
In the case-control study evaluating the micro- tional study estimated the frequency of Campylobacter
biologic profiles of peri-implantitis and healthy peri- rectus, P gingivalis, T forsythia, P intermedia, T denticola,
implant tissues, distinct differences were observed and A actinomycetemcomitans between equivalent
between peri-implantitis and clinically healthy im- peri-implant and periodontal conditions; it showed that
plants.17 Porphyromonas gingivalis, Actinobacillus the occurrence of investigated bacteria was generally
actinomycetemcomitans, Prevotella intermedia, and higher in teeth than in implants.29 The frequency of all
Prevotella nigrescens were identified in 60%, while bacteria except P intermedia was significantly higher in
Staphylococcus epidermidis, enterics, and Candida peri-implantitis compared to healthy implants, while
albicans were seen in 55% of cases. P intermedia and P gingivalis and red-complex species occurred more
P nigrescens were the most common organisms found frequently in peri-implantitis than in peri-implant mu-
in peri-implantitis, while Enterobacter and Klebsiella cositis. P gingivalis and A actinomycetemcomitans were
were the most common of the enterics. However, similarly distributed between periodontitis and peri-
A actinomycetemcomitans was the microorganism most implantitis, but the occurrence of all other species was
significantly associated with peri-implantitis, whereas higher in periodontitis than in peri-implantitis. More-
A actinomycetemcomitans and P gingivalis were seen in over, a case-control study evaluated 78 different bacte-
only one patient with healthy tissues. Another cross- rial species among peri-implantitis and healthy implant
sectional study21 of the microbiologic profiles of 213 samples and showed a bacterial load of T forsythia,
participants with healthy implants, peri-implant mu- P gingivalis, Treponema socranskii, Staphylococcus
cositis, and peri-implantitis demonstrated no signifi- aureus, Streptococcus intermedius, Streptococcus mitis,
cant differences between these conditions. However, and Haemophilus influenzae that was increased by
peri-implant pockets with the deepest probing depth about four times in peri-implantitis when compared to
were correlated with levels of Eikenella corrodens, healthy implants.30 Following comprehensive statisti-
Fusobacterium nucleatum sp vincentii, P gingivalis, and cal analysis, it was suggested that this cluster of bacte-
Micromonas micros. Moreover, the case-control study ria, including T forsythia and S aureus, is associated with
that evaluated the microbiologic profiles of patients peri-implantitis.
with peri-implantitis and healthy subjects did not report Three studies evaluated the association of
higher mean counts of P gingivalis, Treponema denticola, periopathogenic viruses with peri-implantitis.27,28,31
or Tannerella forsythia in both supramucosal and sub- One study27 analyzed the presence of human cyto-
mucosal samples in peri-implantitis patients, whereas megalovirus (HCMV) and Epstein-Barr virus (EBV)
there was no significant difference between supra- and within different peri-implant conditions and showed a
submucosal specimens originating from the same site.22 high prevalence of HCMV and EBV in the subgingival
Another cross-sectional study evaluated periopatho- plaque of peri-implantitis sites. The same group of au-
gens in patients with peri-implantitis, peri-implant mu- thors estimated the prevalence of different genotypes
cositis, and healthy peri-implant tissues and reported of HCMV and EBV in subgingival plaque samples from
high levels of A actinomycetemcomitans, P gingivalis, peri-implantitis, peri-implant mucositis, and healthy
P intermedia, T forsythia, and T denticola in peri-im- implants and reported a high prevalence of HCMV-2
plantitis.19 Another cross-sectional study evaluated 40 and EBV-1 in peri-implantitis.28 Furthermore, an esti-
different bacterial species among patients with peri-im- mation of both periopathogenic bacteria and viruses
plantitis, peri-implant mucositis, and healthy implants in saliva and subgingival samples from healthy implant
and indicated that Actinomyces gerencseriae was pres- and peri-implantitis sites in 23 patients demonstrated
ent in lower mean counts and T forsythia was present in higher counts of EBV, CMV, T denticola, and T forsythia in
higher mean counts in peri-implantitis than in implants peri-implantitis.31 Evaluation of these microorganisms
that were diagnosed with peri-implant mucositis and in as susceptibility factors for peri-implantitis showed

The International Journal of Oral & Maxillofacial Implants 361

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

Table 1 Major Microbiologic Findings Regarding Target Periopathogens in Peri-implantitis


Sampling
Study No. of cases Study method Identification
Leonhardt et al17 (1999) 37 peri-implantitis Case-control PP Culture
51 healthy implants

Mombelli et al18 (2001) 25 peri-implantitis Longitudinal PP Culture, DFM

Hultin et al19 (2002) 17 patients; Cross-sectional PP DNA probe analysis


45 peri-implantitis
53 healthy implants
Leonhardt et al20 (2003) 9 peri-implantitis Longitudinal PP Culture

Renvert et al21 (2007) 31 peri-implantitis Cross-sectional PP DCH


127 peri-implant mucositis
55 healthy implants
Shibli et al22 (2008) 22 peri-implantitis Case-control CT DCH
22 healthy implants
Maximo et al23 (2009) 13 peri-implantitis Cross-sectional CT DCH
12 peri-implant mucositis
10 healthy implants
Tabanella et al24 (2009) 15 peri-implantitis Case-control PP Culture
15 healthy implants (split-mouth)
Persson et al25 (2010) 31 peri-implantitis Randomized PP Expanded DCH assay
longitudinal

Casado et al26 (2011) 10 peri-implantitis, Cross-sectional PP PCR


10 peri-implant mucositis
10 healthy implants
Jankovic et al27 (2011) 20 peri-implantitis Cross-sectional PP Qualitative PCR
18 mucositis
18 healthy implants
Jankovic et al28 (2011) 35 peri-implantitis Cross-sectional PP Qualitative PCR
30 peri-implant mucositis
30 healthy implants
Cortelli et al29 (2013) 53 healthy implants, Cross-sectional CT PCR
53 periodontally healthy
50 peri-implant mucositis
50 peri-implantitis
Persson and Renvert30 (2013) 166 peri-implantitis Case-control PP DCH
47 healthy implants
Verdugo et al31 (2014) 23 peri-implantitis Case-control PP Qualitative PCR for
23 healthy implants (split-mouth) periopathogens,
Quantitative PCR for EBV
and CMV
Sampling: CT = curette; PP = paper points.
Microbiologic methods: DCH = DNA–DNA checkerboard hybridization; DFM = dark-field microscopy.
Bacteria: AA = Aggregatibacter actinomycetemcomitans; AG = Actinomyces gerensceriae; BF = Bacteroides forsythus; CA = Capnocytophaga sp;
CMV = Cytomegalovirus; CR = Campylobacter rectus; EBV = Epstein-Barr virus; EC = Eikenella corrodens; FN = Fusobacterium nucleatum; FU =
Fusobacterium sp; HI = Haemophilus influenzae; HP = Helicobacter pylori; MM = Micromonas micros; PG = Porphyromonas gingivalis; PI = Prevotella
intermedia; PM = Parvimonas micra; PN = Prevotella nigrescens; SA = Staphylococcus aureus; SI = Streptococcus intermedius; SM = Streptococcus
mitis; TF = Tannerella forsythia; TD = Treponema denticola; TS = Treponema socranskii.

that peri-implantitis was 14.2 and 3 times more likely to as well as some opportunistic microorganisms and
harbor EBV than healthy implants and saliva, whereas the responses to different treatment approaches. One
the odds ratios for T denticola and T forsythia were 6.79 study18 included 30 implants affected by peri-implan-
and 3 times higher as well, respectively; thus, EBV was titis and evaluated the treatment effect of tetracycline.
suggested as a potential risk factor for peri-implantitis. At baseline, high frequencies of C rectus, T forsythia,
In the three longitudinal studies considered in this Fusobacterium sp, and P intermedia/nigrescens and low
review,18,20,25 researchers evaluated periopathogens frequencies of A actinomycetemcomitans, P gingivalis,

362 Volume 31, Number 2, 2016

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

Microbiologic findings in peri-implantitis Implant type


Distinct bacterial profile between peri-implantitis and healthy implants. PG, AA, PI, PN in 60%, Brånemark,
Staphylococcus epidermidis, enterics, and Candida albicans in 55%. PI and PN the most common Nobel Biocare
microorganisms, while AA was significantly associated with peri-implantitis. Staphylococci, enterics, and
yeasts more frequent in peri-implantitis than in periodontitis. Similar profile between healthy implants
and periodontium.
45%: CR, FU, PI/PN; TF 36% PI/PN, Fusobacterium sp, BF, and CR reduced levels M1, M3, and M6 ITI
following treatment.
75%–100% implants: AA, FN, PN, PI; 50%–75% implants: PG, PM, CR, EC 14 Brånemark,
3 ITI

7 patients: PI/PN; 6 patients: AA; 3 patients: enterics; 1 patient: PG, SA. PI/PN and enterics persisted 6 Nobel Biocare
mo, 1 y, 5 y posttreatment.
EC, FB sp vincentii, PG, and MM were correlated with the deepest pocket depths. Brånemark,
Nobel Biocare

Higher counts of PG, TD, TF in peri-implantitis. Supra- and subgingival profiles were not substantially Brånemark-like
different.
AG lower counts, TF higher counts in peri-implantitis compared to mucositis and healthy implants. PG Brånemark
was at the highest levels in peri-implantitis. PM, TF, PG, and TD from TS significantly reduced 3 mo
posttreatment in peri-implantitis.
9 implants: FU, TF; 7 implants: CR, PM; 5 implants: PG, PI 11 Brånemark,
4 3i
Baseline: AA (serotype b), FN sp, HP, Staphylococcus sp, and TF. 30 minutes following curettage: reduced No data
counts of AA, Lactobacillus acidophilus, Streptococcus anginosus, and Veillonella parvula. Baseline and 6
mo: no differences in bacterial counts.
AA, PG, PI, TD, and TF identified in all conditions No data

HCMV 13 (65%), EBV in 9 (45%) in peri-implantitis Predominantly


Nobel Biocare

HCMV-2 in 53.3% and EBV-1 in 46.6% peri-implantitis Predominantly


Nobel Biocare

CR, PG, TF, TD, and AA higher in peri-implantitis than in healthy implants. PG and red-complex species Nobel Biocare
higher in peri-implantitis compared to mucositis. PG and AA similar between periodontitis and peri-
implantitis; CR, TF, PI, TD higher in periodontitis.

TF, PG, TS, SA, SI, SM, and HI 4× increased total load in peri-implantitis and suggested a cluster of No data
bacteria, including TF and SA, as related to peri-implantitis.
Higher counts of EBV, CMV, TD, and TF in peri-implantitis. Peri-implantitis 14.2 and 3 times more likely No data
to harbor EBV than healthy implants and saliva, while odds ratios for TD and TF were 6.79 and 3 times
higher than in healthy sites.

and E corrodens were seen.18 Another study20 evaluat- and 1 year posttreatment, P intermedia/P nigrescens
ed the effect of open-flap debridement in conjunction and enterics persisted; after 5 years, P intermedia/
with systemic antibiotics against target microorgan- P nigrescens had reached a peak, as they were now
isms. At baseline, six sites were positive for A actinomy- present in eight patients (versus seven patients at base-
cetemcomitans, seven for P intermedia and P nigrescens, line). Furthermore, in a study that evaluated treatment
one for P gingivalis, one for S aureus, and three for both outcomes in patients with peri-implantitis after two
Escherichia coli and Enterobacter cloacae.20 At 6 months different kinds of mechanical debridement (curettes/

The International Journal of Oral & Maxillofacial Implants 363

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

Table 2 Major Microbiologic Findings Obtained by Metagenomic and Metatranscriptomic


Analysis of the Entire Microbiome in Peri-implantitis
Sampling
Authors N cases Study method Identification
Koyanagi et al32 (2010) 3 peri-implantitis Cross-sectional PP 16S rRNA gene sequencing
3 healthy implants
3 periodontitis

Kumar et al33 (2012) 10 peri-implantitis Cross-sectional PP 16S pyrosequencing


10 healthy implants
10 periodontally healthy
10 periodontitis

Koyanagi et al34 (2013) 6 peri-implantitis, Cross-sectional PP 16S rRNA gene sequencing


6 periodontitis
Tamura et al35 (2013) 15 peri-implantitis Case-control PP Culture +
15 healthy implants 16S DNA gene sequencing

Dabdoub et al36 (2013) 20 peri-implantitis Cross-sectional PP 16S rRNA gene sequencing


20 peri-implant mucositis
13 gingivitis
12 periodontitis
da Silva et al37 (2010) 10 peri-implantitis Case-control CT 16S rRNA gene sequencing
10 healthy implants

Sampling: CT = curette; PP = paper points.


Microbiologic methods: DCH = DNA–DNA checkerboard hybridization; DFM = dark-field microscopy.
Bacteria: AA = Aggregatibacter actinomycetemcomitans; AG = Actinomyces gerensceriae; BF = Bacteroides forsythus; CA = Capnocytophaga sp;
CMV = Cytomegalovirus; CR = Campylobacter rectus; EBV = Epstein-Barr virus; EC = Eikenella corrodens; FN = Fusobacterium nucleatum;
FU = Fusobacterium sp; MM = Micromonas micros; PG = Porphyromonas gingivalis; PI = Prevotella intermedia; PM = Parvimonas micra;
PN = Prevotella nigrescens; SA = Staphylococcus aureus; SM = Streptococcus mutans; TF = Tannerella forsythia; TD = Treponema denticola;
TS = Treponema socranskii.

ultrasound),25 A actinomycetemcomitans (serotype b), F implants and periodontitis. Another cross-sectional


nucleatum sp, Helicobacter pylori, staphylococci, and T study33 analyzed subgingival/submucosal plaque
forsythia were identified at baseline. At 30 minutes af- samples from 40 participants with periodontitis, peri-
ter curettage, the counts of A actinomycetemcomitans, implantitis, and periodontal and peri-implant health
Lactobacillus acidophilus, Streptococcus anginosus, and using 16S pyrosequencing and reported lower lev-
Veillonella parvula were reduced, while at 6 months the els of Prevotella and Leptotrichia sp and higher levels
microflora had been re-established, as evidenced by an of Actinomyces, Peptococcus, and Campylobacter sp,
absence of differences versus the baseline findings. non-mutans streptococci, Butyrivibrio sp, and Strepto-
coccus mutans in peri-implantitis when compared to
Microbiome of Peri-implantitis healthy implants.33 A comparison of peri-implantitis
In a cross-sectional study that analyzed the microbio- and periodontitis demonstrated lower levels of Pre-
logic profile using a sequencing technique, 77 bac- votella sp, non-mutans streptococci, and Lactobacillus,
terial species were identified in peri-implantitis, 57 Selenomonas, Leptotrichia, and Actinomyces sp and higher
in periodontitis, and 12 around healthy implants.32 levels of Peptococcus, Mycoplasma, Eubacterium, Campy-
The microorganisms detected only in peri-implanti- lobacter, and Butyrivibrio sp, S mutans, and Treponema sp
tis were Chloroflexi, Tenericutes, and Synergistetes sp, in peri-implantitis. Hence, the authors concluded that the
P micra, Peptostreptococcus stomatis, Pseudoramibacter microbiomes of periodontitis and peri-implantitis differ
alactolyticus, and Solobacterium moorei, while A ac- significantly. A similar study demonstrated that the micro-
tinomycetemcomitans and P gingivalis were present biologic profile was more diverse in peri-implantitis than
in low levels. Hence, it was concluded that most of in periodontitis.34 Fusobacterium sp and Streptococcus sp
the microbiota in peri-implantitis consisted of gram- were predominant in both disorders, while P micra was
negative species and was more diverse than in healthy detected only in peri-implantitis. This study outlined the

364 Volume 31, Number 2, 2016

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

Microbiologic findings in peri-implantitis Implant type


Peri-implantitis harbored mainly gram-negative species, more diverse microflora than healthy No data
implants and periodontitis. Chloroflexi, Tenericutes, and Synergistetes, PM, Peptostreptococcus
stomatis, Pseudoramibacter alactolyticus, and Solobacterium moorei detected only at peri-implantitis
sites; PG and AA detected at low levels.
Peri-implantitis was associated with lower levels of Prevotella and Leptotrichia and higher levels of Astra Tech, Zimmer,
Actinomyces, Peptococcus, Campylobacter, non-mutans Streptococcus, Butyrivibrio, and SM. Lower Nobel Biocare
levels of Prevotella, non-mutans Streptococcus, Lactobacillus, Selenomonas, Leptotrichia, Actinomyces
and higher levels of Peptococcus, Mycoplasma, Eubacterium, Campylobacter, Butyrivibrio, SM, and
Treponema compared to periodontitis.
The microbial composition of peri-implantitis more diverse than that of periodontitis. Fusobacterium No data
sp and Streptococcus sp were predominant in both diseases. PM detected only in peri-implantitis.
Peri-implantitis about 10-fold higher mean colony-forming units (69 different bacterial species No data
identified) than healthy implants (53 different bacterial species identified). Predominant species
in peri-implantitis: Eubacterium nodatum (7%), PI (5%), FN (3%), Filifactor alocis (3%), E brachy (3%),
Parascardovia denticolens (3%), and PM (3%).
Periodontal microbiome demonstrated significantly higher diversity than the implant; distinct bacterial No data
lineages were associated with health and disease in each ecosystem. Members of the genera
Staphylococcus and Treponema were significantly associated with peri-implantitis.

Great differences were observed between healthy implants compared to peri-implantitis. FN, No data
Campylobacter gracilis, Dialister invisus, Streptococcus sp, Eubacterium infirmum, Filifactor alocis,
and Mitsuokella sp presented a higher mean proportion, while Veillonella dispar, Streptococcus mitis,
Actinomyces meyeri, Granulicatella adiacens showed lower mean proportions in the peri-implantitis
sites than in healthy implants.

higher prevalence of periopathogens and more complex implant sites, where these bacteria were almost un-
microflora in peri-implantitis compared to periodontitis. detectable, it was suggested that this group of bac-
Furthermore, in another case-control study, microbio- teria could play an important role in peri-implantitis.
logic specimens obtained from healthy implant sites and Furthermore, a cross-sectional study was performed36
peri-implantitis were investigated; the results showed to profile peri-implant and periodontal microflora
10-fold higher mean colony-forming units in peri- in healthy and diseased conditions using a sequencing
implantitis.35 The predominant species in peri-implan- method; it revealed that peri-implant and periodontal
titis sites were Streptococcus and Eubacterium, while microbiomes are distinct ecosystems and indicated
the predominant species around healthy implants a more diverse profile in periodontitis. Members of
were Streptococcus, Veillonella, and Actinomyces. Al- the genera Staphylococcus and Treponema were sig-
though Streptococcus was common in both groups, nificantly associated with peri-implantitis.36 A case-
the prevalence of bacterial species was completely control study that estimated microbiomes of healthy
different between the investigated groups. Sixty-nine implants and peri-implantitis showed different profiles
different bacterial species were identified in peri- for peri-implantitis and healthy implants; peri-implantitis
implantitis sites, and predominant bacterial species was associated with more periopathogenic bacte-
were Eubacterium nodatum, E brachy, E saphenum, Fi- rial species than healthy implants.37 Higher mean
lifactor alocis, Slackia exigua, Parascardovia denticolens, proportions of F nucleatum, Campylobacter gracilis,
P intermedia, F nucleatum, P gingivalis, Centipeda peri- Dialister invisus, Streptococcus sp, human oral taxon (HOT)
odontii, and P micra; the number of species at healthy 064, Eubacterium infirmum, F alocis, and Mitsuokella sp
implants was 53. Based on the significantly higher HOT 131, along with lower mean proportions of Veillon-
prevalence of nonsaccharolytic anaerobic gram-pos- ella dispar, S mitis, Actinomyces meyeri, and Granulicatella
itive rods in peri-implantitis and compared to healthy adiacens, were the findings in peri-implantitis.

The International Journal of Oral & Maxillofacial Implants 365

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

DISCUSSION periodontal infection.42 This hypothesis indicates that


similar bacteria are present in both healthy and dis-
As pointed out in a previous review of this issue,38 early eased conditions, while the changes in bacterial pro-
studies focused on comparisons of the microbiologic portions represent the key pathologic determinant.
profiles in peri-implantitis and chronic periodontitis In addition, it is suggested that the most important
using routine culture techniques, hybridization, and role of periopathogens is the conversion of a symbi-
PCR to evaluate target periopathogenic bacteria, ie, otic ecosystem into a dysbiotic one.43 Translated to
the so-called periopathogens. In brief, these studies peri-implantitis, it seems that periopathogens provide
showed that the qualitative profile of periopathogens permissive conditions for the overgrowth of opportu-
was similar between healthy and peri-implantitis– nistic bacteria, thus orchestrating their conversion from
affected implants, whereas the quantitative composi- physiologically harmless to pathological members of
tion of periopathogens was the distinguishing factor microflora. In this regard, an evaluation of opportunis-
between health and disease. Furthermore, an analysis tic bacteria is the subject of recent studies that focused
of the periopathogen profiles of healthy versus dis- primarily on the identification of these bacteria. One
eased implants and corresponding conditions in teeth study30 quantitatively evaluated opportunistic bacteria
revealed an increased frequency of periopathogens together with periopathogens and estimated their risk
in teeth. Generally, investigations of periopathogens rate. It showed that, of 78 analyzed bacteria, a cluster
were unable to establish a clear microbiologic pro- of four opportunistic bacteria (S aureus, S intermedius,
file associated with peri-implantitis, and it was ac- S mitis, and H influenzae) and three periopathogens
cepted that the microbiologic and immunopathologic (T forsythia, P gingivalis, and T socranskii) are associated
profiles of peri-implantitis and periodontitis differ.5 with peri-implantitis.30 This additionally supports the
Therefore, more recent studies have been directed theory that opportunistic bacteria and their quantita-
toward comprehensive analyses of the microbiome tive content represent the important determinants of
in peri-implantitis.33–37 Advanced molecular tech- the microbiologic profile in peri-implantitis.
niques, such as sequencing methods, render a com- Considering the reviewed studies as a whole, there
prehensive overview of phylogenetic and taxonomic is a high rate of inhomogeneity in their protocols that
bacterial diversity and indirectly compensate for the significantly reduces the applicability of the reported
limitations of cultures and lack of reactivity of con- results. The use of several different diagnostic protocols
ventional biochemical tests that are typical for a num- contributed to inhomogeneity in the expression of mi-
ber of fastidious anaerobic microorganisms.39 For the crobiologic outcome variables (total bacterial counts,
aforementioned reasons, during the last 4 years the frequencies, proportions, loads, etc); thus, compari-
newer sequencing approaches have been widely used sons of the findings between groups remain limited.
to profile the peri-implantitis microflora and offer new In general, the reviewed studies were performed on
insights. Microbiologic screens employing sequencing relatively small samples (never more than 50 peri-
techniques have shown the following: (1) peri-implan- implantitis cases, and, on average, 20 peri-implantitis
titis harbored mainly gram-negative species, and an cases) and provided very heterogenous observational
association of certain periopathogens with peri-im- findings that render between-study comparisons dif-
plantitis was confirmed; (2) nonsaccharolytic bacteria ficult. Hence, despite the great efforts of the implant
are associated with peri-implantitis; (3) S aureus is not research community, most of the reported results are
strictly present in peri-implantitis; (4) a different and observational and descriptive, such that it is almost im-
more diverse microflora is present in peri-implantitis possible to determine meaningful patterns concerning
than in healthy implants; (5) the microbiomes in peri- the microbiologic profile of peri-implantitis. Further-
implantitis and periodontitis are different ecosystems. more, earlier studies mostly evaluated the qualitative
In fact, these metagenomic and metatranscriptomic and quantitative profiles of periopathogens, whereas
techniques revealed more diverse microbiologic pro- more recent studies revealed the implications of oth-
files in both periodontitis and peri-implantitis than er pathogens, which remain merely identified, while
previously thought, and in general it has been sug- their quantitative profiles and pathologic mechanisms
gested that the role of periopathogens must be re- have not yet been clearly established. Moreover, the
interpreted.40 When considering the reported results reported results originated predominantly from case-
as a whole, the peri-implantitis microflora consists of control and cross-sectional studies (only 3 of 21 were
periopathogens and opportunistic bacteria in almost longitudinal studies), although it seems that changes
the same measure.41 The lack of conclusive evidence in bacterial counts represent the key determinant of
about the impact of periopathogens on the peri-im- peri-implantitis microflora. Therefore, future research
plantitis microflora could be possibly explained by the in this area should be oriented toward the quantita-
recently reported “keystone pathogen” hypothesis of tive assessment of peri-implant microflora cohabitants

366 Volume 31, Number 2, 2016

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

in well-designed prospective studies. Since it is obvi- ACKNOWLEDGMENTS


ous that the peri-implantitis microbiologic profile is
complex and determined by numerous members with The authors report no conflicts of interest related to this study.
complex interrelationships, advanced statistical tools for
risk estimation should be considered in future research
as well. REFERENCES
When considering the microbiologic profile from
a clinical standpoint, peri-implantitis is associated 1. Albrektsson T, Isidor E. Consensus report of session IV. In: Lang NP,
with aggressive and resistant bacterial strains and vi- Karring T (eds). Proceedings of the First European Workshop on
Periodontology. London: Quintessence, 1994:365–369.
ruses; therefore, mechanical anti-infective treatment 2. Mombelli A, Lang NP. The diagnosis and treatment of peri-implan-
usually fails to reduce or eliminate periopathogens titis. Periodontol 2000 1998;17:63–76.
without auxiliary antimicrobial therapy.18,25,44 Further- 3. Renvert S, Persson GR. Periodontitis as a potential risk factor for
peri-implantitis. J Clin Periodontol 2009;36(suppl 10):9–14.
more, the opportunistic bacteria characteristic of peri- 4. Mouhyi J, Dohan Ehrenfest DM, Albrektsson T. The peri-implanti-
implantitis are highly resistant and do not respond to tis: Implant surfaces, microstructure, and physicochemical aspects.
some antibiotics that are routinely administered in peri- Clin Implant Dent Relat Res 2012;14:170–183.
5. Berglundh T, Zitzmann NU, Donati M. Are peri-implantitis le-
odontology, such as metronidazole. Additionally, it was sions different from periodontitis lesions? J Clin Periodontol
previously reported that the implant-abutment connec- 2011;38(suppl 11):188–202.
tion could harbor a reservoir of bacteria in both healthy 6. Ericsson I, Berglundh T, Marinello C, Liljenberg B, Lindhe J. Long-
standing plaque and gingivitis at implants and teeth in the dog.
and diseased implants, based on the “circular model” of Clin Oral Implants Res 1992;3:99–103.
bacterial contamination.45,46 Therefore, it seems that the 7. Marinello CP, Berglundh T, Ericsson I, Klinge B, Glantz PO, Lindhe J.
contaminated implant surface represents a reservoir of Resolution of ligature-induced peri-implantitis lesions in the dog.
J Clin Periodontol 1995;22:475–479.
periopathogens with the ability to firmly adhere to both 8. Toijanic JA, Ward CB, Gewerth ME, Banakis ML. A longitudinal
biotic and abiotic systems.47 This could possibly explain clinical comparison of plaque-induced inflammation between
the better treatment outcome in patients in whom sur- gingival and peri-implant soft tissues in the maxilla. J Periodontol
2001;72:1139–1145.
face decontamination was performed25 compared to 9. Gualini F, Berglundh T. Immunohistochemical characteristics of
the outcome following routine mechanical treatment inflammatory lesions at implants. J Clin Periodontol 2003;30:14–18.
with curettes and ultrasound devices.23 Related to this, 10. Rakic M, Lekovic V, Nikolic-Jakoba N, Vojvodic D, Petkovic-Curcin A,
Sanz M. Bone loss biomarkers associated with peri-implantitis. A
microbiologic contamination of the implant surface cross-sectional study. Clin Oral Implants Res 2013;24:1110–1116.
represents an intrinsic characteristic of peri-implant pa- 11. Heitz-Mayfield LJ, Lang NP. Comparative biology of chronic and
thology that can interrupt the stability of the implant aggressive periodontitis vs. peri-implantitis. Periodontol 2000
2010;53:167–181.
surface oxide layer and stimulate further detrimental 12. Albrektsson T, Dahlin C, Jemt T, Sennerby L, Turri A, Wennerberg
processes. Finally, effective infection control through A. Is marginal bone loss around oral implants the result of a
targeted anti-infective approaches, implant surface provoked foreign body reaction? Clin Implants Dent Relat Res
2014;16:155–165.
decontamination, and supportive periodontal therapy 13. Roccuzzo M, Bonino F, Aglietta M, Dalmasso P. Ten-year results of a
remains crucial treatment for peri-implantitis. three arms prospective cohort study on implants in periodontally
compromised patients. Part 2: Clinical results. Clin Oral Implants
Res 2012;23:389–395.
14. Mombelli A, Lang NP. The diagnosis and treatment of periimplanti-
CONCLUSIONS tis. Periodontol 2000 1998;17:63−76.
15. Tonetti M. Risk factors for osseodisintegration. Periodontol 2000
1998;17:55–62.
Considering the reviewed studies as a whole, the mi- 16. Khan KS, ter Riet G, Popay J, Nixon J, Kleijen J. Stage II: Conducting
crobiologic profile in peri-implantitis: (1) is complex the review. Phase 5: Study quality assessment. In: Khan KS, ter Riet
and variable, (2) consists of gram-negative anaerobic G, Glanville J, Sowden AJ, Kleijnen J (eds). Undertaking Systematic
Reviews of Research on Effectiveness, ed 2. York, University of
periopathogens and opportunistic microorganisms York, 2001:1–20.
in almost the same ratio, (3) is frequently associated 17. Leonhardt A, Renvert S, Dahlén G. Microbial findings at failing
with Epstein-Barr virus and nonsaccharolytic anaerobic implants. Clin Oral Implants Res 1999;10:339–345.
18. Mombelli A, Feloutzis A, Brägger U, Lang NP. Treatment of peri-im-
gram-positive rods, (4) is not so strictly associated with plantitis by local delivery of tetracycline. Clinical, microbiological
Staphylococcus aureus, and (5) is different from that of and radiological results. Clin Oral Implants Res 2001;12:287–294.
periodontitis. It seems that the presence of titanium cre- 19. Hultin M, Gustafsson A, Hallstrom H, Johansson LA, Ekfeldt A,
Klinge B. Microbiological findings and host response in patients
ates a distinct microenvironment, and as a consequence, with peri-implantitis. Clin Oral Implants Res 2002;13:349–358.
the microbiologic profile in peri-implantitis remains 20. Leonhardt A, Dahlén G, Renvert S. Five-year clinical, microbiologi-
different from that of periodontitis. Furthermore, micro- cal, and radiological outcome following treatment of peri-implan-
titis in man. J Periodontol 2003;74:1415–1422.
organisms that constitute the microbiologic profile in 21. Renvert S, Roos-Jansaker AM, Lindahl C, Renvert H, Persson GR.
peri-implantitis have the potential to disrupt the stability Infection at titanium implants with or without a clinical diagnosis
of the implant surface layer and, hence, might contrib- of inflammation. Clin Oral Implants Res 2007;18:509–516.

ute to a pathologic interface with the implant surface.

The International Journal of Oral & Maxillofacial Implants 367

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.
Rakic et al

22. Shibli JA, Melo L, Ferrari DS, Figueiredo LC, Faveri M, Feres M. Com- 35. Tamura N, Ochi M, Miyakawa H, Nakazawa F. Analysis of bacterial
position of supra- and subgingival biofilm of subjects with healthy flora associated with peri-implantitis using obligate anaerobic
and diseased implants. Clin Oral Implants Res 2008;19:975–982. culture technique and 16S rDNA gene sequence. Int J Oral Maxillofac
23. Maximo MB, de Mendonc AC, Santos VR, de Figueiredo LC, Feres Implants 2013;28:1521–1529.
M, Duarte PM. Short-term clinical and microbiological evaluation 36. Dabdoub SM, Tsigarida AA, Kumar PS. Patient-specific analysis of
of peri-implant diseases before and after mechanical anti-infective periodontal and peri-implant microbiomes. J Dent Res 2013;92:
therapies. Clin Oral Implants Res 2009;20:99–108. 168S–175S.
24. Tabanella G, Nowzari H, Slots J. Clinical and microbiological 37. da Silva ES, Feres M, Figueiredo LC, Shibli JA, Ramiro FS, Faveri
determinants of ailing dental implants. Clin Implant Dent Relat Res M. Microbiological diversity of peri-implantitis biofilm by Sanger
2009;11:24–36. sequencing. Clin Oral Implants Res 2013;25:1–8.
25. Persson GR, Samuelsson E, Lindahl C, Renvert S. Mechanical non- 38 Mombelli A, Decaillet F. The characteristics of biofilms in peri-im-
surgical treatment of peri-implantitis: A single-blinded randomized plant disease. J Clin Periodontol 2011;38(suppl 11):203–213.
longitudinal clinical study. II. Microbiological results. J Clin Periodon- 39. Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Defining
tol 2010;37:563–573. the normal bacterial flora of the oral cavity. J Clin Microbiol
26. Casado PL, Otazu IB, Balduino A, de Mello W, Barboza EP, Duarte ME. 2005;43:5721–5732.
Identification of periodontal pathogens in healthy periimplant sites. 40. Hajishengallis G. The inflammophilic character of the periodontitis-
Implant Dent 2011;20:226–235. associated microbiota. Mol Oral Microbiol 2014;29:248–257.
27. Jankovic S, Aleksic Z, Dimitrijevic B, Lekovic V, Camargo P, Kenney 41. Belibasakis GN. Microbiological and immuno-pathological aspects
B. Prevalence of human cytomegalovirus and Epstein-Barr virus in of peri-implant diseases. Arch Oral Biol 2014;59:66–72.
subgingival plaque at peri-implantitis, mucositis and healthy sites. 42. Hajishengallis G, Darveau RP, Curtis MA. The keystone-pathogen
A pilot study. Int J Oral Maxillofac Surg 2011;40:271–276. hypothesis. Nat Rev Microbiol 2012;10:717–725.
28. Jankovic S, Aleksic Z, Dimitrijevic B, Lekovic V, Milinkovic I, Kenney B. 43. Hajishengallis G, Lambris JD. Microbial manipulation of receptor
Correlation between different genotypes of human cytomegalovi- crosstalk in innate immunity. Nat Rev Immunol 2011;11:187–200.
rus and Epstein-Barr virus and peri-implant tissue status. Aust Dent J 44. Persson GR, Salvi GE, Heitz-Mayfield LJA, Lang NP. Antimicrobial
2011;56:382–388. therapy using a local drug delivery system (Arestins) in the treat-
29. Cortelli SC, Cortelli JR, Romeiro RL, et al. Frequency of periodontal ment of peri-implantitis. I: Microbiological outcomes. Clin Oral
pathogens in equivalent peri-implant and periodontal clinical Implants Res 2006;17:386–393.
statuses. Arch Oral Biol 2013;58:67–74. 45. Canullo L, Penarrocha-Oltra D, Soldini C, Mazzocco F, Penarrocha
30. Persson GR, Renvert S. Cluster of bacteria associated with peri- M, Covani U. Microbiological assessment of the implant-abutment
implantitis. Clin Implant Dent Relat Res 2014;16:783–793. interface in different connections: Cross-sectional study after 5 years
31. Verdugo F, Castillo A, Castillo F, Uribarri A. Epstein-Barr virus as- of functional loading. Clin Oral Implants Res 2015;26:426–434.
sociated peri-implantitis: A split-mouth study. Clin Oral Investig 46. Canullo L, Peñarrocha-Oltra D, Covani U, Orlato Rossetti PH. Micro-
2015;29:535–543. biological and clinical findings of implants in healthy condition and
32. Koyanagi T, Sakamoto M, Takeuchi Y, Ohkuma M, Izumi Y. Analysis with peri-implantitis. Int J Oral Maxillofac Implants 2015;30:834–842.
of microbiota associated with peri-implantitis using 16S rRNA gene 47. McAlister AD, Sroka A, Fitzpatrick RE, et al. Gingipain enzymes from
clone library. J Oral Microbiol 2010;2:5104–5110. Porphyromonas gingivalis preferentially bind immobilized extracellu-
33. Kumar PS, Mason MR, Brooker MR, O’Brien K. Pyrosequencing lar proteins: A mechanism favouring colonization? J Periodontal Res
reveals unique microbial signatures associated with healthy and 2009;44:348–353.
failing dental implants. J Clin Periodontol 2012;39:425–433.
34. Koyanagi T, Sakamoto M, Takeuchi Y, Maruyama N, Ohkuma M, Izumi
Y. Comprehensive microbiological findings in peri-implantitis and
periodontitis. J Clin Periodontol 2013;40:218–226.

368 Volume 31, Number 2, 2016

© 2016 BY QUINTESSENCE PUBLISHING CO, INC. PRINTING OF THIS DOCUMENT IS RESTRICTED TO PERSONAL USE ONLY.
NO PART MAY BE REPRODUCED OR TRANSMITTED IN ANY FORM WITHOUT WRITTEN PERMISSION FROM THE PUBLISHER.

You might also like