You are on page 1of 31

Periodontology 2000, Vol. 71, 2016, 82–112 © 2016 John Wiley & Sons A/S.

& Sons A/S. Published by John Wiley & Sons Ltd


Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Antibiotics/antimicrobials:
systemic and local
administration in the therapy of
mild to moderately advanced
periodontitis
KARIN JEPSEN & SØREN JEPSEN

Periodontitis is a plaque-induced inflammatory con- and limitations of both forms of administration are
dition that affects the periodontium; it is caused by listed in Table 1. Systemic delivery has the potential
the adherence to tooth surfaces of pathogenic bacte- advantage of reaching pathogens widely distributed
rial species organized in complex communities that in the oral cavity, including those in nondental oral
form biofilms (129, 130). Standard periodontal treat- niches, such as the dorsum of the tongue and the
ment consists of mechanical debridement to remove crypts of the tonsils. However, it requires high patient
biofilm and calculus from the affected root surfaces. compliance, can lead to unwanted systemic side
This approach has proven successful for most effects affecting compliance and may contribute to
patients (48, 49, 154, 175). However, mechanical bacterial resistance (160, 189, 228). In contrast, local
debridement is a highly demanding procedure with administration is independent of patient compliance
some limitations, such as the inability to access deep and allows the application of drugs to the site of
pockets, surface irregularities and furcation areas infection at a concentration that cannot be reached
(104). Because of the infectious nature of periodonti- by the systemic route. However, their application is
tis, the rationale for using adjunctive antimicrobial limited to isolated clinically detected lesions, and
agents is to eradicate or reduce the numbers of reinfection from nontreated sites or nondental oral
pathogenic bacteria in deep pockets, root furcations niches may occur.
and concavities or those residing at or within the This review aims to update the current knowledge
periodontal tissues at the biofilm–gingival interface. lt of adjunctive antimicrobial therapy of periodontitis
is hoped that pathogenic organisms which are not based on the most recent scientific evidence. Special
accessible to mechanical removal by hand or power- emphasis is given to the implications for mild and
driven instruments can be reduced in number or moderate forms of the disease. Mild to moderately
eliminated by antimicrobial therapy (220). Biofilm- advanced periodontitis refers to findings from the
associated infections in general are difficult to treat clinical examination and is generally characterized by
with antibiotics unless the biofilm is disrupted periodontal probing depths of up to 6 mm with clini-
mechanically, as in anti-infectious periodontal ther- cal attachment loss of up to 4 mm. Signs and symp-
apy (129, 130). Therefore, the current general consen- toms of edema, erythema, gingival bleeding upon
sus is that if antimicrobial therapy is considered, it probing and/or suppuration may be present and
should be preceded by thorough mechanical debride- associated with increased tooth mobility as well as
ment to disrupt the structured biofilm (66). radiographic evidence of bone loss (16–30% or
Adjunctive antimicrobial therapy can be delivered >3 mm but ≤5 mm) (1, 10). The effects of standard
either systemically or locally. The possible advantages nonsurgical periodontal therapy with regard to

82
Adjunctive antibiotic/antimicrobial therapy in periodontitis

Table 1. Comparison of local and systemic antimicrobial therapy

Source: Mombelli (139).

pocket reduction and attachment Ievel gain for the zole plus amoxicillin achieved the best results but the
different categories of initial periodontal probing aspects related to appropriate dosage and duration
depth are well documented and can serve as a bench- remained unclear. Herrera et al. (94) updated their
mark against which the possible additional effects of previous work to include 32 publications and 45 com-
adjunctive antimicrobials can be compared. parisons. The consensus paper of the corresponding
workshop for this paper (176) concluded that, if sys-
temic antimicrobials were used, this should be as part
Effects of adjunctive systemic of nonsurgical periodontal therapy. Indirect evidence
antibiotics suggests that antibiotic intake should start on the day
of debridement completion and be completed within
A wide range of systemic antibiotics, including amoxi- a short period of time. Furthermore, because of prob-
cillin (with or without clavulanic acid), azithromycin, lems related to the indiscriminate use of antimicro-
clindamycin, doxycycline, metronidazole, spiramycin bials (i.e. systemic side effects, adverse effects and
and tetracycline, have been tested in clinical studies. increase in bacterial resistance), the use of systemic
The effects of such adjunctive systemic antimicrobial antimicrobials in periodontitis should be restricted to
therapy in chronic and aggressive periodontitis have patients with aggressive, severe and progressing
been evaluated in a number of reviews and system- forms of periodontitis. Keestra et al. (112, 113) pub-
atic reviews with meta-analyses (57, 58, 66, 84, 94–96, lished two systematic reviews and meta-analyses on
112, 113, 161). Herrera et al. (96) performed a meta- the adjunctive use of systemic antibiotics in patients
analyses of 25 clinical trials (randomized controlled with untreated chronic periodontitis (43 randomized
trials and controlled clinical trials) of ≥6 months’ controlled trials) and aggressive periodontitis (14 ran-
duration, comparing subgingival debridement (scal- domized controlled trials). The authors reported sig-
ing and root planing) with adjunctive systemic antibi- nificant additional pocket depth reduction with the
otics vs. scaling and root planing alone or with use of antibiotics for either condition of periodontal
placebo. The authors reported statistically significant disease. Statistically, no specific type of antibiotic was
additional benefits for spiramycin (probing-depth superior over another. Separate meta-analyses were
change = 0.41 mm) and amoxicillin plus metronida- performed for moderately deep and deep pockets. A
zole (clinical attachment level change = 0.45 mm) in recent systematic review published by Garcia Canas
deep pockets (>6 mm). They concluded that in speci- et al. (66) was designed as an update of the system-
fic clinical situations, such as patients with deep atic review conducted by Herrera et al. (96) and
pockets or with progressive ‘active’ disease or with selected 23 clinical trials published after 2001 with a
specific profiles, use of adjunctive systemic antimi- minimum of 6 months’ follow up and patients diag-
crobials could be clinically relevant. Haffajee et al. nosed with chronic periodontitis or aggressive peri-
(84) published a systematic review including 29 stud- odontitis. Overall, a tendency toward improvement
ies (26 randomized controlled trials and three cohort was found in studies in which systemic antibiotics
studies) with a duration of more than 1 month. were used as adjunctive therapy with scaling and root
Periodontal conditions were aggressive, chronic or planing, but because of the high level of heterogene-
recurrent periodontitis, as well as periodontal ity of the studies the authors could not establish
abscesses. The authors concluded that metronida- definitive conclusions for chronic periodontitis and
zole, tetracycline and the combination of metronida- guidelines regarding the use of adjunctive systemic

83
Jepsen & Jepsen

antibiotics. Therefore, clinicians should make their mean differences were small. In another systematic
treatment decisions for each patient individually after review, Zandbergen et al. (238) analyzed 28 clinical
weighing the benefits and the risks. trials (including 19 randomized controlled trials) on
the effect of concomitant administration of amoxi-
cillin plus metronidazole adjunctive to scaling and
Amoxicillin and metronidazole
root planing over at least 1 month in patients with
In recent years, the antibiotic therapy most widely aggressive or chronic periodontitis. The authors con-
documented in clinical reports has been the combina- cluded that the clinical benefits of nonsurgical peri-
tion of metronidazole and amoxicillin (190, 191, 238). odontal therapy can be enhanced by adjunctive
Sgolastra et al. (190, 191) published two systematic systemic antimicrobial therapy in adults who are
reviews on the efficacy of adjunctive amoxicillin plus otherwise healthy. Rabelo et al. (161) performed a sys-
metronidazole: one in patients with chronic periodon- tematic review and a Bayesian network meta-analysis
titis (four randomized controlled trials) and the other to assess the effects of adjunctive systemic antibiotics
in patients with aggressive periodontitis (six random- in aggressive periodontitis. They found that for clinical
ized controlled trials). All patients had been treated attachment gain/reduction in probing depth, and
with scaling and root planing, alone, or in combina- compared with scaling and root planing alone, the
tion with amoxicillin and metronidazole. The results combinations of scaling and root planing + metron-
of the metaanalysis in patients with aggressive peri- idazole (mean difference = 1.08 mm/1.05 mm) and
odontitis, in which full-mouth data were analyzed, scaling and root planing + metronidazole + amoxi-
showed significantly more clinical attachment gain cillin (mean difference = 0.45 mm/ 0.53 mm) had the
(0.4 mm) and probing depth reduction (0.6 mm) after most beneficial outcomes.
full-mouth scaling and root planing in combination
with systemic administration of these two antibiotics.
Effects of systemic antibiotics in initially
Patients with chronic periodontitis also seemed to
moderate or deep periodontal sites
experience a clinical benefit of this antibiotic combi-
nation compared with scaling and root planing alone. To put the observed weighted mean differences of
The results of the meta-analyses showed significantly 1.4 mm for pocket depth reduction and 0.9 mm for
more mean clinical attachment gain (0.2 mm) and clinical attachment level gain into perspective, and
probing depth reduction (0.4 mm) after adjunctive because not all studies included had a comparative
amoxicillin and metronidazole, although the addi- treatment arm of scaling and root planing only, Zand-
tional mean benefit was less distinct than in the bergen et al. (238) made an indirect comparison with
patient group with aggressive forms of the disease data originating from another review published by
(Table 2). Therefore, these systematic reviews, based one of the co-authors (35) by analyzing the overall
on full-mouth clinical data, seem to support a statisti- effect of nonsurgical therapy alone. In this explora-
cal benefit for the adjunctive use of amoxicillin plus tory analysis, data stratified according to baseline
metronidazole in the treatment of chronic and aggres- pocket depth were compared with data previously
sive periodontitis; however, the observed weighted reported by Cobb (34), for nonsurgical debridement

Table 2. Therapeutic efficacy of nonsurgical periodontitis treatment, with and without adjunctive systemic amoxicillin
and metronidazole, in patients with chronic periodontitis and aggressive periodontitis

From Sgolastra et al. (190, 191). Follow up varied from 3 to 24 months.

84
Adjunctive antibiotic/antimicrobial therapy in periodontitis

antibiotics. Patients who received antibiotics at initial sites improving above clinically relevant thresholds,
therapy showed statistically significant improvement compared with patients who received antibiotics at
in pocket depth reduction and in the percentage of retreatment in sites with remaining pockets ≥5 mm.

A D

B E

C F

93
Adjunctive antibiotic/antimicrobial therapy in periodontitis

3. Aimetti M, Romano F, Torta I, Cirillo D, Caposio P, 21. Boyton RJ, Reynolds CJ, Quigley KJ, Altmann DM. Immune
Romagnoli R. Debridement and local application of tetra- mechanisms and the impact of the disrupted lung micro-
cycline-loaded fibres in the management of persistent biome in chronic bacterial lung infection and bronchiecta-
periodontitis: results after 12 months. J Clin Periodontol sis. Clin Exp Immunol 2013: 171: 117–123.
2004: 31: 166–172. 22. Brusca SB, Abramson SB, Scher JU. Microbiome and
4. Akalin FA, Baltacioglu E, Sengu € n D, Hekimoglu S, Taskin mucosal inflammation as extra-articular triggers for
M, Etikan I, Fisenk I. A comparative evaluation of the clini- rheumatoid arthritis and autoimmunity. Curr Opin
cal effects of systemic and local doxycycline in the treat- Rheumatol 2014: 26: 101–107.
ment of chronic periodontitis. J Oral Sci 2004: 46: 25–35. 23. Cahenzli J, Ko € ller Y, Wyss M, Geuking MB, McCoy KD.
5. Al-Joburi W, Quee TC, Lautar C, Iugovaz I, Bourgouin J, Intestinal microbial diversity during early-life colonization
Delorme F, Chan ECS. Effects of adjunctive treatment of shapes long-term IgE levels. Cell Host Microbe 2013: 14:
periodontitis with tetracycline and spiramycin. J Periodon- 559–570.
tol 1989: 60: 533–539. 24. Cani PD. Gut microbiota and obesity: lessons from the
6. Aminov RI. The role of antibiotics and antibiotic resistance microbiome. Brief Funct Genomics 2013: 12: 381–387.
in nature. Environ Microbiol 2009: 11: 2970–2988. 25. Caricilli A, Saad M. The role of gut microbiota on insulin
7. Angaji M, Gelskey S, Nogueira-Filho G, Brothwell D. A sys- resistance. Nutrients 2013: 5: 829–851.
tematic review of clinical efficacy of adjunctive antibiotics 26. Carlet J. World alliance against antibiotic resistance: the
in the treatment of smokers with periodontitis. J Periodon- WAAAR declaration against antibiotic resistance. Med
tol 2010: 81: 1518–1528. Intensiva 2014: 39: 34–39; http://dx.doi.org/10.1016/
8. Antunes LCM, Han J, Ferreira RBR, Lolic P, Borchers CH, j.medin.2014.10.004.
Finlay BB. Effect of antibiotic treatment on the intestinal 27. Carvalho J, Novak MJ, Mota LF. Evaluation of the effect of
metabolome. Antimicrob Agents Chemother 2011: 55: subgingival placement of chlorhexidine chips as an
1494–1503. adjunct to scaling and root planing. J Periodontol 2007: 78:
9. Ardila CM, Granada MI, Guzma n IC. Antibiotic resistance 997–1001.
of subgingival species in chronic periodontitis patients. J 28. Casarin RC, Peloso Ribeiro ED, Sallum EA, Nocviti FH Jr,
Periodontal Res 2010: 45: 557–563. Goncßalves RB, Casati MZ. The combination of amoxicillin
10. Armitage GC. Development of a classification system for and metronidazole improves clinical and microbiologic
periodontal diseases and conditions. Ann Periodontol results of one-stage, full-mouth, ultrasonic debridement in
2000: 4: 1–6. aggressive periodontitis treatment. J Periodontol 2012: 83:
11. Armitage GC. Learned and unlearned concepts in peri- 988–998.
odontal diagnostics: a 50-year perspective. Periodontol 29. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife
2000 2013: 62: 20–36. WT, Schmidt TM, Young VB. Decreased diversity of the
12. Azmak DN, Atilla G, Luoto H, Sorsa T. The effect of subgin- fecal microbiome in recurrent Clostridium difficile-associ-
gival controlled-release delivery of chlorhexidine chip ated diarrhea. J Infect Dis 2008: 197: 435–438.
onclinical parameters and matrix metalloproteinase-8 levels 30. Chassaing B, Gewirtz AT. Gut microbiota, low-grade
in gingival crevicular fluid. J Periodontol 2002: 73: 603–615. inflammation, and metabolic syndrome. Toxicol Pathol
13. Baltacioglu E, Aslan M, Sarac O, Saybak A, Yuva P. Analysis 2014: 42: 49–53.
of clinical results of systemic antimicrobials combined 31. Cionca N, Giannopoulou C, Ugolotti G, Mombelli A.
with nonsurgical periodontal treatment for generalized Amoxicillin and metronidazole as an adjunct to full-mouth
aggressive periodontitis: a pilot study. J Can Dent Assoc scaling and root planing of chronic periodontitis. J Peri-
2011: 77: b97. odontol 2009: 80: 364–371.
14. Baquero F. Metagenomic epidemiology: a public health 32. Cionca N, Giannopoulou C, Ugolotti G, Mombelli A.
need for the control of antimicrobial resistance. Clin Microbiologic testing and outcomes of full-mouth scaling
Microbiol Infect 2012: 18: 67–73. and root planing with or without amoxicillin/metronida-
15. Becker W, Becker BE, Ochsenbein C, Kerry G, Caffesse R, zole in chronic periodontitis. J Periodontol 2010: 81: 15–23.
Morrison EC, Prichard J. A Longitudinal study comparing 33. Citorik RJ, Mimee M, Lu TK. Bacteriophage-based syn-
scaling, osseous surgery and modified Widman proce- thetic biology for the study of infectious diseases. Curr
dures. J Periodontol 1987: 59: 351–365. Opin Microbiol 2014: 19: 59–69.
16. Belda-Ferre P, Alcaraz LD, Cabrera-Rubio R, Romero H, 34. Cobb CM. Non-surgical pocket therapy. Ann Periodontol
Simo  n-Soro A, Pignatelli M, Mira A. The oral metagenome 1996: 1: 443–490.
in health and disease. ISME J 2011: 6: 46–56. 35. Cobb CM. Clinical significance of non-surgical periodontal
17. Bergstro € m J. Tobacco smoking and risk for periodontal therapy: an evidence-based perspective of scaling and root
disease. J Clin Periodontol 2003: 30: 107–113. planing. J Clin Periodontol 2002: 29(Suppl 2): 6–16.
18. Blaser M. Antibiotic overuse: stop the killing of beneficial 36. Costello EK, Stagaman K, Dethlefsen L, Bohannan B, Rel-
bacteria. Nature 2011: 476: 393–394. man DA. The application of ecological theory towards an
19. Bogren A, Teles RP, Torresyap G, Haffajee AD, Socransky understanding of the human microbiome. Science 2012:
SS, Wennstrom JL. Locally delivered doxycycline during 336: 1255–1262.
supportive periodontal therapy: a 3-year study. J Periodon- 37. Cosyn J, Wyn I, De Rouck T, Sabzevar MM. A chlorhexidine
tol 2008: 79: 827–835. varnish implemented treatment strategy for chronic peri-
20. Bonito DAJ, Lux L, Lohr KN. Impact of local adjuncts to odontitis. J Clin Periodontol 2005: 32: 750–756.
scaling and root planing in periodontal disease therapy: a 38. Cosyn J, Wyn I, De Rouck T, Sabzevar MM. Clinical bene-
systematic review. J Periodontol 2005: 76: 1227–1236. fits of subgingival chlorhexidine varnish application as an

105
Table 4. Evaluation of initially deep sites in five studies: probing-depth reduction and clinical attachment gain were determined in test subjects after treatment of gen-
eralized aggressive periodontitis using scaling and root planing in conjunction with amoxicillin + metronidazole compared with placebo control

Initially deep sites (probing depth varied from ≥6 mm to ≥7 mm).



Mann–Whitney U-test.

From Mestnik et al. (136).
*Bonferroni-corrected t-test.
**Student’s t-test.
***Kruskall–Wallis test.
****Mann–Whitney U-test.
*****Analysis of covariance.
******One-way analysis of variance P-value.
Adjunctive antibiotic/antimicrobial therapy in periodontitis

87
Jepsen & Jepsen

Table 5. Mean reduction of pocket depth and gains in syndromes that substantially modify susceptibility
clinical attachment following scaling and root planing and/or progression of disease (11, 89, 98, 202). An
example of a patient with generalized severe peri-
odontitis treated with adjunctive systemic antibiotics
in a specialist practice is shown in Fig. 2(A–H).

Effects of adjunctive locally


delivered antimicrobials
Various antimicrobials and delivery systems have
been developed with the aim to minimize systemic
uptake of the antimicrobial agent whilst maintain-
ing high levels in the crevicular fluid at the target.
These agents, which provide substantivity of an
antimicrobial system at an effective drug concentra-
*From Hung & Douglass (100). References from which the results were calcu-
lated were 5, 15, 40, 122, 126, 142, 155, 156. Mean numbers, weighted according tion over time, have been investigated mostly in
to sample size, after scaling and root planing were analyzed using baseline localized forms of periodontitis and in nonrespond-
measures of initial probing depth as covariant.
ing, persistent and recurrent sites (60, 114, 221). Sci-
entific evidence using adjunctive locally delivered
deep sites. For patients diagnosed with mild to mod-
antibiotics is currently based on three systematic
erate periodontitis (1), the majority of probing depths
reviews and one meta-analysis (20, 86, 100, 133).
recorded at baseline usually do not exceed 6 mm.
Hanes & Purvis (86) published a systematic review
The effects of mechanical debridement alone are well
of 19 studies that evaluated the efficacy of different
documented for moderately deep sites (4–6 mm),
nonsurgical periodontal therapies, including admin-
and clinicians can expect a mean reduction in pocket
istration of local sustained-release agents (minocy-
depth of approximately 1 mm and a gain in clinical
cline gel, microencapsulated minocycline,
attachment level of approximately 0.5 mm (100)
chlorhexidine chip and doxycycline gel). In a subse-
(Table 5). Thus, the risk is low for residual deep sites
quent systematic review, the statistically significant
(>5 mm) with persistence of high loads of bacterial
adjunctive benefit for adjunctive tetracycline,
pathogens, the limited additional benefit of the use of
minocycline, metronidazole and chlorhexidine
systemic antibiotics in moderately deep sites does not
could be confirmed with modest clinical improve-
justify their use. Exceptions may be very rare cases of
ments in probing depth reductions compared with
young patients with moderate disease who are
scaling and root planing alone (20).
infected with the JP2 genotype of Aggregatibacter acti-
A recent systematic review (133) was based on data
nomycetemcomitans (Fig. 11A–G); This distinct geno-
extracted from 52 randomized controlled trials,
type of A. actinomycetemcomitans has been found to
reported in 56 publications. In most of the studies,
be strongly associated with aggressive forms of peri-
site selection for the application of antimicrobial
odontitis, particularly in patients from African medi-
delivery systems was based on the presence of sites
terranean regions (e.g. Morocco), but is not
with pockets of >5 mm in patients with chronic peri-
associated with known systemic conditions or genetic

Fig. 1. (A) A 23-year-old patient from North Africa observed and plaque index was 54%. (E) Explanation of
unhappy with her smile line. Her maxillary incisors periodontal status. (F) Re-evaluation 8 weeks after com-
showed distinct migration (flaring) with incomplete lip pletion of debridement on two consecutive days with
closure. This patient has localized periodontitis not associ- adjunctive systemic azithromycin; periodontal conditions
ated with known systemic conditions or genetic are clearly improved. Azithromycin was used because of its
syndromes. (B) Clinical view at baseline: distinct black- potent activity against periodontal pathogens, short treat-
stained calculus on the lower right canine. (C) Radio- ment regimen (ensuring good patient compliance), low
graphic documentation; upper incisors show moderate incidence of side effects, persisting antibiotic effects and
bone loss. (D) Periodontal attachment status and plaque even longer-lasting immunomodulatory effects after a sin-
index at baseline. Probing depths ranged from 2 mm to 6 gle course (97). (G) Clinical view 8 weeks after nonsurgical
mm and in localized areas (e.g. upper right wisdom tooth) therapy. Fig. 1 (A), (B) and (G) are photographs courtesy of
were up to 9 mm, generalized attachment loss was T. Waller.

88
Adjunctive antibiotic/antimicrobial therapy in periodontitis

odontitis. The overall meta-analysis, combining all with the control groups. Therefore, through subgingi-
antimicrobial products, showed significant pocket val application of an antimicrobial adjunct, an addi-
depth reductions and gains in clinical attachment tional clinical benefit in reductions of pocket probing
level (0.4 and 0.3 mm, respectively) when compared depth could be demonstrated (Table 6a) (133). The

A B

89
Jepsen & Jepsen

Fig. 2. (A) Clinical status of a 48-year-old patient with sev- otics (amoxicillin 500 mg, three times daily for 8 days; and
ere generalized periodontitis, showing distinct signs of metronidazole 400 mg twice daily for 8 days). (F) Peri-
acute infections on the upper incisors and lower left odontal status 6 months after debridement and adjunctive
canine. (B) Radiographs of the same patient showing mod- systemic antibiotics; the bleeding score is 8%. (G) Clinical
erate-to-severe bone loss. (C) Periodontal status before status 6 months after localized corrective surgery as well
treatment, with pocket depth up to 12 mm and a bleeding as supportive periodontal therapy, presenting a bleeding
score of 100%. (D) Explanation of periodontal status. (E) score of 4%. (H) Periodontal status 6 months after local-
Clinical status 6 months after completion of debridement ized corrective surgery as well as supportive periodontal
on two consecutive days with adjunctive systemic antibi- therapy; the bleeding score is 4%.

largest effect was demonstrated with the application Antibiotics, in addition to sonic/ultrasonic subgingi-
of tetracycline fibers, resulting in a pocket depth val instrumentation, were applied to sites with
reduction of 0.7 mm, followed by doxycycline pocket depth ≥5 mm. The authors concluded that
(0.5 mm pocket depth reduction) and minocycline topically administered slow-release doxycycline gel
(0.4 mm pocket depth reduction). The effect of may provide a short-term benefit in controlling
chlorhexidine chips and metronidazole rendered inflammation and deep pockets in treated periodon-
minimal additional probing improvements, below tal patients participating in a secondary prevention
0.4 mm. Regarding clinical attachment level gain, the program and able to maintain a satisfactory Ievel of
highest effect (0.9 mm) was demonstrated by the oral hygiene. Bogren et al. (19) studied a total of 128
application of chlorhexidine–xanthan gel, although periodontal maintenance patients, with at least four
these data are based on only one study (133) teeth with pocket depth ≥5 mm treated with local
(Table 6b). Other antimicrobials yielded additional application of doxycycline gel, at baseline and at 1
clinical attachment level gains of 0.2–0.6 mm. How- and 2 years, as an adjunct to mechanical debride-
ever, the application of metronidazole and other ment or mechanical debridement only. The authors
chlorhexidine products did not add any benefit over concluded that although short-term effects on clini-
scaling and root planing alone. In spite of no signifi- cal parameters were found with the adjunctive use of
cant changes in bleeding, and a high heterogeneity of locally delivered doxycycline, applications repeated
data, Matesanz-Pe rez et al. (133) concluded that annually had no clinical effects beyond those
scientific evidence supports the adjunctive use of observed with mechanical debridement alone in
local antimicrobials to debridement in deep or recur- maintenance patients. Tomasi et al. (207) studied 22
rent periodontal sites, mostly when using vehicles patients with chronic periodontitis, who received ini-
with proven sustained release of the antimicrobial. tial root debridement by ultrasonic instrumentation,
An example of a tooth treated with adjunctive locally followed by random assignment to retreatment of
delivered antibiotics as part of the initial treatment residual furcation pockets at 3 months by ultrasonic
phase is shown in Fig. 3(A–G). instrumentation, with or without adjunctive local
application of a 8.8% doxycycline gel, and concluded
that the improvement in molar furcation involve-
Local adjunctive antimicrobials for
ment after nonsurgical periodontal therapy was not
recurrent or persistent periodontal lesions
enhanced by adjunctive locally applied doxycycline.
Although the probability of observing a high percent- An example of a patient treated with adjunctive
age of deep residual pockets after anti-infective ther- locally delivered antibiotics for recurrent disease is
apy in patients with mild to moderate periodontitis is shown in Fig. 4(A–H).
low (Table 5), sites with pocket depth >5 mm repre-
sent an incomplete periodontal treatment outcome
and require further therapy (134). In particular,
bleeding sites, which carry an increased risk of dis- Treatment protocols for adjunctive
ease progression, are difficult to control. Renvert & antibiotics
Persson (167) reported that the presence of deep
residual pockets after treatment was associated with
Time of administration
further disease progression. Tonetti et al. (209) evalu-
ated the efficacy of a slow-release doxycycline gel Griffiths et al. (76), in a follow-up study of a random-
adjunctively administered with nonsurgical therapy ized controlled trial performed by Guerrero et al. (80)
in subjects with recurrent or persistent periodontitis. evaluated whether retreatment with adjunctive

90
Adjunctive antibiotic/antimicrobial therapy in periodontitis

A B

91
Jepsen & Jepsen

Table 6. Clinical therapeutic efficacy of scaling and root planing vs. scaling and root planing plus different local
antimicrobials on (a) probing pocket depth and (b) clinical attachment level in patients with chronic periodontitis

From Matesanz-P
erez et al. (133). Follow up varied from 6 months to more than 12 months.

metronidazole and amoxicillin would give the original 2 months and the two study groups were crossed-
placebo group benefits comparable with the original over for treatments. The test group was treated with
test group. The previous study was extended for debridement and the control group with systemic

Fig. 3. (A) Frontal view of a 52-year-old man with an acute of inflammatory reactions are still evident in the lower
periodontal lesion on an upper left incisor and generalized incisors. (E) Re-evaluation 8 weeks after root surface
moderate periodontitis (from (106) Quintessence with per- debridement using piezo-surgical instruments (EMS,
mission). (B) Periodontal status before treatment: the Electro Medical Systems, Nyon, Switzerland) and adminis-
probing depth ranged from 2 to 6 mm (localized up to tration of local antibiotics (Arestinâ, OraPharma)
10 mm on the upper-left central incisor), the plaque index subgingivally on the upper left central incisor. Probing
was 100% and the bleeding score was 73%. (C) Radio- depths on that tooth were still between 3 and 10 mm but
graphs of the same patient showing generalized slight to there were no visible signs of suppuration. (F) Clinical sta-
moderate bone loss and the upper left central incisor tus: stable condition 2 years after regenerative procedures
showing severe bone loss. (D) Clinical status 8 weeks after and one year after orthodontics. (G) Periodontal status
administration of local antibiotics (Arestinâ; OraPharma, 3 years after regenerative procedures on the upper left
Horsham, PA, USA), gentle debridement of the upper left central incisor and 3-monthly supportive periodontal
central incisor and nonsurgical periodontal therapy. Areas therapy.

92
Adjunctive antibiotic/antimicrobial therapy in periodontitis

antibiotics. Patients who received antibiotics at initial sites improving above clinically relevant thresholds,
therapy showed statistically significant improvement compared with patients who received antibiotics at
in pocket depth reduction and in the percentage of retreatment in sites with remaining pockets ≥5 mm.

A D

B E

C F

93
Jepsen & Jepsen

A C H

E
D

Fig. 4. (A) Clinical status of a 65-year-old otherwise sys- (Arestinâ; OraPharma, Inc.). Before surgery there was no
temically healthy patient receiving supportive periodontal suppuration, but pocket depth remained unchanged. (E)
therapy. Recurrent disease with evidence of suppuration is Evidence of a large intrabony defect to the mesial of the
apparent on the lower right canine. (B) Two periodontal lower canine. Monophilic sutures in place after treating the
examinations carried out 3 months apart. Sudden peri- defect with enamel matrix proteins and bone replacement
odontal breakdown the lower right canine, full-mouth pla- material (Emdogainâ, Straumann; Bioossâ, Geistlich,
que/bleeding score: 10%/12%. (C) Results of a microbial Switzerland). (F) Clinical situation 1 year after regenerative
test taken from the subgingival biofilm of the canine, surgery. (G) Radiographs taken at patient0 s first visit/exam-
including a susceptibility profile to different antibiotic sub- ination with an unexpected periodontal breakdown 2 years
stances. (D) Situation after root surface debridement and later. Radiographic defect fill 1 year after regenerative sur-
application of adjunctive minocycline microspheres gery. (H) Examination 1 year after regenerative surgery.

In deep pockets (≥7 mm), the mean difference was reductions, and changes in the number of pockets
0.9 mm (P = 0.003) and in moderate pockets (4– that were in need of further treatment at 2, 6 and 8
6 mm) it was 0.4 mm (P = 0.036) (Table 7a). Clini- months, are presented in Table 7b. For pockets con-
cally relevant changes of >2 mm in probing depth verting from ≥5 mm to ≤4 mm, the difference was

94
Adjunctive antibiotic/antimicrobial therapy in periodontitis

83% for initial antibiotic treatment compared with dontal therapy. They performed a randomized con-
67% for delayed antibiotic treatment (P = 0.041); and trolled crossover clinical trial and investigated the
for pockets converting from ≥4 mm to ≤3 mm, the adjunctive effect of systemic antibiotics on the
difference was 63% for initial antibiotic treatment nonsurgical and surgical treatment phases. The
compared with 49% for delayed antibiotic treatment authors reported similar long-term results, but stated
(P = 0.297). The authors concluded that: ‘patients that antibiotic therapy during the first nonsurgical
that received the antibiotics at the initial therapy treatment phase resolved the disease more quickly
showed significant additional benefits compared to and thus reduced the need for additional surgical
those who received the same regime at the re-treat- intervention.
ment phase.’ A limitation of the study design was dif-
ferent follow-up times of 6 months from initial
Systemic antibiotics and full-mouth
antibiotic treatment vs. 2 months for delayed
disinfection
antibiotic treatment. Very recently, Mombelli et al.
(140) investigated the possible benefits of amoxicillin In patients with generalized aggressive periodontitis,
and metronidazole in the different phases of perio- the clinical and microbiological effects of a full-

Table 7. (a) Improvements in pocket-depth reduction and in the percentage of sites above clinically relevant thresh-
olds, compared with patients who received antibiotics at retreatment in sites with remaining pockets ≥5 mm; (b) tim-
ing of systemic antimicrobials (metronidazole and amoxicillin) given at either initial therapy or in the retreatment
phase in patients with generalized aggressive periodontitis

From Griffiths et al. (76).


*Analysis of covariance for probing pocket depth reduction and gain in lifetime cumulative attachment loss.
**Subset analysis showing clinically relevant changes in probing pocket depth presented at median and interquartile range (IQ) using the Mann–Whitney U-test.

95
Jepsen & Jepsen

mouth disinfection protocol comprising debride- A prospective multicenter randomized controlled


ment combined with chlorhexidine irrigation, rins- trial designed to investigate the course of peri-
ing and tongue brushing, with or without amoxicillin odontal disease and the long-term impact of
plus metronidazole, were studied (92, 214). Although adjunctive systemic antibiotics was performed by
some short-term advantages were observed for clini- Harks et al. (87). Four-hundred and six patients
cal variables, both clinical and microbiological with moderate-to-severe periodontitis received
observations suggested an important impact, in both nonsurgical periodontal therapy and adjunctive
groups, of the therapy provided; however, after systemic antibiotics (amoxicillin plus metronida-
6 months there were no relevant differences zole) and maintenance therapy at 3-month inter-
between groups with or without the additional vals. After a 27.5-month observation period, both
adjunctive antibiotic therapy. On the other hand, treatments were found to be effective in preventing
Aimetti et al. (2) assessed the adjunctive effect of disease progression (the percentage of sites show-
amoxicillin and metronidazole over a complete full- ing further attachment loss was ≥1.3 mm). The
mouth disinfection protocol, and both clinical and median percentage of sites showing further attach-
microbiological findings revealed significant benefits ment loss was 7.8% in the placebo group
of adjunctive antibiotics after 6 months. In patients compared with 5.3% in the group receiving adjunc-
with chronic periodontitis, no differences in clinical tive antibiotics. Empiric therapy with adjunctive
attachment level gain or pocket depth reduction systemic antibiotics showed a small absolute,
were found when a full-mouth disinfection protocol although statistically significant, additional reduc-
with metronidazole was compared with the same tion of further attachment loss.
protocol with placebo (158).

Smokers
Smokers are at a greater risk of developing periodon-
Best maintained clinical results over time
tal disease and tooth loss compared with nonsmokers
A study designed to find a periodontal treatment (17, 146, 203, 204) and respond less favorably to peri-
with the best clinical results that were maintained odontal treatment (92, 108, 146). In general, smoking
over 24 months, as evaluated by changes in pocket reduced clinical responses to all therapies applied,
depth and clinical attachment level, was performed namely nonsurgical, surgical and adjunctive therapies
by Goodson et al. (71). Treatment outcomes of (71). In the maintenance phase of periodontal ther-
scaling and root planing, alone, or in combination apy, heavy smoking was one of the main risk factors
with systemic antibiotics, local antibiotic therapy for disease progression (134). Most of the clinical tri-
and/or periodontal surgery, were compared under als addressing post-therapy microbiological changes
a common protocol. The authors concluded that agree that it is more difficult to control subgingival
patients receiving adjunctive therapies generally pathogens by mechanical periodontal therapy in
exhibited improved attachment gain and/or prob- smokers than in nonsmokers (43, 78, 131, 212, 227).
ing depth reduction when compared with debride- This is probably because of the reduced capacity of
ment alone. Adjunctive locally delivered smokers to combat periodontal pathogens (81), as it
tetracycline had no statistically significant effects. has been demonstrated that smoking affects the cel-
Adjunctive combination of amoxicillin and metron- lular and humoral inflammatory responses (172).
idazole provided additional benefits regarding Therefore, adjunctive therapeutic approaches have
attachment level gain and pocket reduction. Peri- been suggested for treating periodontitis in smokers.
odontal surgery provided pocket reduction, but was Superior clinical outcomes have been reported in
not associated with attachment level gain (Table 8). smokers compared with nonsmokers, with nonsurgi-
This study was conducted with a factorial design to cal treatment using adjunctive systemic or locally
test whether or not treatments interact with each applied antimicrobial agents (115, 132, 198, 206). For
other. The test of interaction between therapies persistent pockets of >5 mm after a course of debride-
(cross products) for 24-month clinical attachment ment, Kinane & Radvar (115) concluded that smoking
level gain and probing pocket depth reduction may have an important role in determining the prog-
revealed no statistical significance. The investigators nosis of periodontal treatment. Treatment outcomes
therefore concluded that among adjunctive thera- may be improved by application of locally delivered
pies, an additive effect, but no synergistic or antag- antimicrobial systems as adjuncts to scaling and root
onistic impact, was observed. planing. Tomasi et al. (206) studied 103 patients (42

96
Adjunctive antibiotic/antimicrobial therapy in periodontitis

Table 8. Clinical attachment level gain and probing pocket depth reduction, from baseline to 24 months, in sites with
pocket depth >5 mm at baseline, of patients randomized to eight treatment groups

From Goodson et al. (71).


Data were evaluated using analysis of covariance with treatment as the dependent variable in the eight groups; a matrix of 56 pairwise comparison probabilities was
obtained. Attachment level gain and pocket depth reduction values are given as adjusted means  standard error of the mean, with the same superscript letter
within each column indicating a significant difference at P < 0.0009; significant pairwise differences were determined by post-hoc analysis using Fisher’s Protected
Least Significant Difference test.

smokers and 61 nonsmokers), each having at least microbiota are obtained with the adjunctive use of
eight periodontal sites with probing pocket depth of metronidazole and amoxicillin (Table 9). However, a
≥5 mm, treated with root planing or ultrasonic instru- systematic review concluded that the evidence for an
mentation and application of an 8.5% doxycycline gel. additional benefit of adjunctive antibiotic therapy in
The authors concluded that locally applied antibiotics smokers with chronic periodontitis is insufficient and
may partly counteract the negative effect of smoking inconclusive. Additional well-designed randomized
on periodontal healing following nonsurgical therapy. controlled trials are required to assess the effect of
Matarazzo et al. (132) evaluated the clinical and antibiotics in conjunction with periodontal treat-
microbiological effects of scaling and root planing, ments in smokers (7). Thus, from an overall health-
alone, or combined with metronidazole or with care perspective it can be argued whether it is reason-
metronidazole and amoxicillin, in the treatment of able to compensate for the reduced treatment
smokers with chronic periodontitis over a short time response in smokers by the use of antibiotics or
period, and found that the greatest benefits in clinical whether these patients should be involved in a smok-
parameters and in the composition of the subgingival ing-cessation program (165).

Table 9. Treatment effect in smokers with chronic periodontitis: probing-depth reduction and clinical attachment
gain (pre- and post-treatment) following scaling and root planing, in conjunction with the combination of amoxicillin
and metronidazole, compared with placebo controls, after 3 months

From Matarazzo et al. (132).


*Kruskal–Wallis/Mann–Whitney U-test.

97
Jepsen & Jepsen

Patients with type II diabetes significant improvement in the glycemic parameters


at any time point (Table 10b).
Miranda et al. (138) studied the effects of adjunctive
systemic amoxicillin and metronidazole in 58 patients
with uncontrolled type II diabetes and generalized Patients with specific microbiologic
chronic periodontitis. The group receiving debride- profiles
ment in combination with antibiotics showed greater As a result of its proven ability to suppress A. actino-
mean probing depth reductions and clinical attach- mycetemcomitans in periodontitis lesions and other
ment gains (Table 10a), as well as a lower number of oral sites (69), the combination of amoxicillin and
sites with probing depths of ≥5 mm as the primary metronidazole appears well suited, especially to treat
outcome variable and a reduced number of subjects A. actinomycetemcomitans-associated periodontitis.
with nine or more of these residual pockets compared Van Winkelhoff et al. (229) reported on the microbio-
with the control group up to 1 year post-treatment logical and clinical effects of mechanical debride-
(P < 0.05). The authors concluded that the adjunctive ment, in combination with metronidazole plus
use of amoxicillin and metronidazole significantly amoxicillin therapy, in patients with A. actino-
improved the clinical outcomes of debridement alone mycetemcomitans-associated periodontitis. The
in the treatment of patients with type II diabetes and results showed that the combined treatment was very
chronic periodontitis. Despite the clinical improve- effective in suppressing subgingival A. actino-
ments obtained with the adjunctive use of antimicro- mycetemcomitans in patients with severe periodonti-
bials, none of the therapies yielded statistically

Table 10. (a) Treatment effect in patients with uncontrolled type II diabetes and chronic periodontitis: probing-depth
reduction and clinical attachment gain (pre- and post-treatment) following scaling and root planing in conjunction
with the combination of amoxicillin and metronidazole compared with placebo controls after 1 year; (b) Changes in
glycated hemoglobin and fasting plasma glucose with time and according to treatment

From Miranda et al. (138).


Values are given as mean  standard deviation.
Changes within groups in glycated hemoglobin and fasting plasma glucose levels over time were tested by repeated-measures analysis of variance (P > 0.05).
P-values were calculated using analysis of covariance.
*Differences between groups were tested using the Student’s t-test (P > 0.05).

98
Adjunctive antibiotic/antimicrobial therapy in periodontitis

tis, but the variation in clinical treatment responses tions. Another complication may be that even com-
between the patients could not be correlated with the mensal bacteria can trigger an immune response if
microbial flora. Dannewitz et al. (41) evaluated the they reach critically high numbers or are present in
clinical and microbiological effects of the combina- an inappropriate habitat within the host.
tion of mechanical and antibiotic periodontal therapy
in subjects who tested positive for subgingival A. acti-
Morbidity/adverse events for systemic
nomycetemcomitans. At an average follow up of
and local antibiotics
39 months with an antibiotic regime of either amoxi-
cillin/metronidazole or ciprofloxacin/metronidazole, Nausea, vomiting and gastrointestinal discomfort are
the clinical situation had improved significantly in all the most common adverse events reported in studies
subjects after systemic periodontal therapy and on the use of adjunctive systemic antibiotics in
remained stable during the course of the postopera- patients with periodontitis (28, 31, 32, 52, 53, 61, 71,
tive follow up. No differences in the clinical data were 79, 80, 121, 132, 136, 168, 194, 233). Other reported
found between the subjects that tested positive and adverse effects include headaches (31, 32, 92, 214), a
negative for A. actinomycetemcomitans in the postop- metallic taste (31, 32, 92, 194, 214), general malaise
erative period. Mombelli et al. (141) studied the (79, 80) or a burning sensation of the mouth or ton-
effects of adjunctive amoxicillin in patients with mod- gue (92, 214). Other side effects include muscu-
erate to advanced periodontitis, with or without loskeletal and respiratory disorders (31, 32), dry
A. actinomycetemcomitans. No specific benefits of the mouth, erythema oral ulceration, dizziness, staining
antibiotics were seen in patients positive for A. acti- of tongue or teeth (92, 214), irritability (194), a rash on
nomycetemcomitans. Oteo et al. (148) investigated the face or neck and nausea after the use of alcohol
systemic azithromycin, as an adjunct to debridement, (61). Griffiths et al. (76) reported a high incidence of
in Porphyromonas gingivalis-positive patients with adverse events, with 42% of subjects affected
moderate chronic periodontitis. This systemic antimi- (Table 11). The majority of these side effects were
crobial was chosen because of its convenient dosage, considered to be minor, but serious adverse events,
and the results showed significant benefits in both sufficient to withdraw the medication, occurred in
clinical and microbiological outcome variables after two patients. One subject developed a severe rash
6 months. These results, however, were not corrobo- and the other had to discontinue the medication
rated in a similar study, in which patients were not because of nausea, diarrhea and drowsiness. For most
selected based on a specific microbiological profile, published reports and studies on locally administered
and where no adjunctive effect was observed at the 1- antibiotics, no significant patient-centered adverse
year evaluation (174). Altogether the studies showed events have been reported (20, 86). However, gingival
inconclusive results. One reason for this may be vari- redness, pain on the first day of local administration,
ations in the susceptibility profiles of these clinically gingival tingling, fever, headache, diarrhea, periodon-
relevant oral pathogens, of which knowledge is criti-
cal to provide the patient with the appropriate antibi- Table 11. Adverse events reported following antibiotic
otic therapy. treatment in a study with 19 patients

Side effects of antibiotics


The side effects and adverse reactions related to the
use of antibiotics in the treatment of periodontitis, or
of infectious diseases in general, must always be
weighed against the possible benefits. Antibiotic-
induced pharmacotoxic adverse reactions are listed
in the summary of characteristics of every product
and should be taken into account. In addition, the
antiinflammatory or immune-suppressive effects of
antibiotics, able to mimic a successful therapy out-
come of the infection, are not well understood. The
intake of multiple antibiotics at the same time leaves
From Griffiths et al. (76).
a patient susceptible to development of yeast infec- Values are given as n (%).

99
Jepsen & Jepsen

tal abscesses, root sensitivity, taste disturbances and nate use of antimicrobials (149) and has generated
stomatitis have been noted in a recent systematic great concern among health authorities. Whilst the
review (133). discovery of antibiotics 70 years ago has revolution-
ized medicine, this increase of antibiotic resistance in
bacteria is the subject of intense debate driven by
Impact of antibiotic therapy on the
economic and political concerns. In nature0 s complex
gastrointestinal microbiome
ecosystem, antibiotics are found in multiple environ-
Antibiotics have changed the way we are treating ments, fauna and flora. Researchers discovered that
infectious diseases, but we are just beginning to microbes produced metabolites with antibiotic activ-
assess the collateral damage on the symbiotic ity mainly in low concentrations, serving primarily as
microorganisms living in the gastrointestinal tract signaling molecules for bacterial cooperation and
(18). Repeated exposure to broad-spectrum antibi- only became antimicrobial at much higher concen-
otics dramatically alters the composition of human trations (62). Studies have also shown that ‘antibiotic
gut microbial communities, which may persist for resistance is ancient’ (44) and much larger numbers
long periods of time (46). Experiments in mice have of antibiotic-resistance genes are naturally present in
demonstrated that antibiotic exposure disrupts human commensal microbial genomes, as well as in
intestinal homeostasis and, in addition, has a pro- soil samples throughout the world, than previously
found impact on the intestinal metabolome, affecting recognized (42, 199) Remarkably, resistance has also
the levels of over 87% of all detectable metabolites been detected to antibiotics that have only been
(8). The consequences of such microbial shifts in the recently introduced for clinical use (e.g. telithomycin
intestine can alter the proper maturation of mucosal and tigecycline) (44). Therefore, the reservoir of
immunity and render individuals more susceptible to antibiotic-resistance genes has definitely increased
infection (186). Antibiotic-associated diarrhea caused since the discovery of antibiotics more than 70 years
by Clostridium difficile, reflecting colonization of a ago (118). The global pandemic of antibiotic resis-
disrupted microbial community with a specific tance (Fig. 5) represents a major health and eco-
microbe, can cause severe intestinal illness with nomic burden (Fig. 6), and, together with a relative
potentially life-threatening complications (29). lack of innovation in generating new antibiotics, can
The current view of the impact of antimicrobial only be solved if scientists, politicians, society and
treatment on the gastrointestinal microbiota is that business work together, nationally and internation-
culture-independent studies may show persistent ally, pursuing diverse, coordinated approaches. In the
changes in the microbiota that last for years (36, 106). present day, pathogens can spread quickly through
The clinical significance of these changes is currently the globalized system of travel, trade and food distri-
unknown, but there is growing evidence that chronic bution, presenting a threat to the entire world (171,
diseases are linked to dysbiosis of the intestinal 210). We have to realize the danger of returning to a
microbiota. These include asthma and allergic dis- pre-antibiotic era, with the risk of many infectious
eases (99, 218), autoimmune diseases (22, 23), type 1 diseases becoming untreatable and uncontrollable.
diabetes (211, 223), type 2 diabetes (25, 240), obesity The problem of antibiotic resistance is also of great
(24, 120, 137), metabolic syndrome (30, 50), chronic concern to the dental profession and has been
disorders of the gut (51, 111), atherosclerosis (74), addressed by Walker et al. (219, 220). To date, most
undernutrition (72, 117, 119), celiac disease (157, 187) periodontal antibiotic treatment regimens appear to
and chronic lung infections (21, 45). In addition, have been empirically prescribed without guidance
many investigators have proposed that the vast gath- from a microbiologic analysis of the subgingival bac-
ering of microflora in our intestines could have a terial biofilm populations (56). Patients with peri-
major impact on our state of mind. To paraphrase odontitis frequently yield multiple species of
one of these opinions regarding this link: ‘The micro- periodontal pathogens that may vary in their degree
biome may yield a new class of psychobiotics for the of resistance to antibiotics (197). To provide a patient
treatment of anxiety, depression and other mood dis- with an appropriate antibiotic therapy, it may be criti-
orders’ (185). cal to know the susceptibility profiles of clinically rel-
evant oral pathogens. The oral microbiota also seems
to be an important reservoir for transferable antimi-
Bacterial resistance
crobial resistance (166, 170, 216, 217). Approximately
The development of bacterial resistance has 50% of the Enterococcus faecalis strains isolated from
increased dramatically as a result of the indiscrimi- patients with marginal and apical periodontitis were

100
Adjunctive antibiotic/antimicrobial therapy in periodontitis

Fig. 5. Available national data on resistance for nine selected bacterial/antibacterial drug combinations, 2013 (Source:
World Health Organization, with permission).

Fig. 6. Estimates of burden of antibacterial resistance (Source: World Health Organization, with permission).

101
Jepsen & Jepsen

found to be resistant to one or more of the antimicro- therapeutic potential of single-drug regimens with
bial agents tested, mostly tetracycline and/or ery- beta-lactam antibiotics in periodontal therapy. The
thromycin (164). A description of resistance in the in vitro effectiveness of metronidazole against nearly
oral biofilm as a result of horizontal gene transfer was all beta-lactamase-producing subgingival bacterial
reported in vivo, when Streptococcus cristaceus species recovered further supports clinical periodon-
acquired a transposon that conferred doxycycline titis-treatment strategies involving the combination
resistance from a strain of Streptococcus oralis. Both of systemic amoxicillin plus metronidazole. Recently,
strains were isolated from the subgingival biofilm of Rams et al. (163) investigated subgingival biofilm
patients undergoing doxycycline therapy as part of specimens from inflamed deep periodontal pockets
their periodontal treatment (224). Recently, a taken from 400 adults with chronic periodontitis in
plasmidome within E. faecalis, isolated from patients the USA. Overall, 74.2% of the patients were reported
with marginal periodontitis in Norway, was charac- to have subgingival periodontal pathogens resistant
terized and the authors concluded that the majority to at least one of the antibiotics commonly used in
of E. faecalis strains in marginal periodontitis lesions clinical periodontal practice. The authors concluded
are likely to be a reservoir for diverse mobile genetic that the wide variability found in periodontal patho-
elements and associated antimicrobial resistance gen antibiotic-resistance patterns should concern
determinants (200). clinicians empirically selecting antibiotic treatment
In northern European countries, where antibiotic regimens for patients with chronic periodontitis.
usage is generally restricted and infrequent, antibiotic
resistance was reported to be rare among periodontal
pathogens tested from patients with periodontitis
(215). This is in contrast to southern Europe, Colum- Future challenges and
bia and South America, where access to antibiotics is considerations
less stringently controlled and there is more non-
supervised consumption of these drugs, and where a In the coming years there will be major conceptual
clearly higher level of periodontal pathogen antibiotic changes in health and disease diagnostics followed by
resistance has been found (9, 228). Veloo et al. (215) appropriate treatment concepts. Microorganisms
investigated the antibiotic-susceptibility profiles of inhabit almost every imaginable environment in our
five periodontal pathogens (Prevotella intermedia, biosphere and play an integral and unique role in all
Fusobacterium nucleatum, A. actinomycetemcomi- ecosystems. We have learned that there are thou-
tans, P. gingivalis and Parvimonas micra) to six com- sands of microbial strains and species associated with
monly used antibiotics in periodontics using the the human body, contributing significantly more
European Committee on Antimicrobial Susceptibility genes than the host genome itself. Their structure,
Testing and Clinical and Laboratory Standards Insti- function, interaction and dynamics are critical to our
tute breakpoints for interpretation of the results. In existence, and intense research efforts have been
comparison with a previous study from the Nether- made to understand the impact of the human micro-
lands (227) minor differences in susceptibility profiles biome on health and disease (16, 75, 152, 195). Signifi-
were noted, but the minimum inhibitory concentra- cant progress has been made in the development and
tions for amoxicillin, clindamycin, azithromycin and application of high-throughput technologies for ana-
tetracycline were higher for 90% of A. actinomycetem- lyzing microbial community structure and functions.
comitans strains. In general, geographical differences Complex analysis systems are transforming our ability
were found in the susceptibility profiles of P. gingi- to investigate the composition of microbial commu-
valis and A. actinomycetemcomitans between Euro- nities with its full spectrum of host–microbe interac-
pean countries. A comparison of susceptibility tions (241). Many challenges still remain, although
profiles between European subjects and Colombian metagenome-specific assembly algorithms (159) and
subjects revealed a much higher level of resistance in methods for ‘binning’ genomes from metagenome
the latter. data (192, 231) have led to numerous successes, and
Rams et al. (162) studied subgingival plaque speci- the current stage of cataloging and functional analy-
mens from deep periodontal pockets of 564 adults ses will continue to progress with DNA and RNA
with severe chronic periodontitis before treatment. sequencing.
Fifty-two per cent of the patients yielded betalacta- In the case of periodontitis, dysbiotic and highly
mase-producing species. The authors concluded that organized climax biofilm communities resist host
this finding would raise questions about the elimination and have optimal conditions for growth

102
Adjunctive antibiotic/antimicrobial therapy in periodontitis

in a nutritionally favorable inflammatory environ- tant for developing new therapies for disrupting inter-
ment. Mature subgingival biofilms may have actions between major oral pathogens.
immune-subversive and pro-inflammatory roles, Host-modulation strategies involving anti-inflam-
thereby causing collateral damage to the periodon- matory strategies are another approach to inhibit
tium. The magnitude of this damage is host depen- destructive inflammation by antimicrobial effects
dent, relative to its genetic predisposition, and through limiting inflammatory exudate-derived nutri-
environmental factors may also play a role (85). A ents or blocking immune-evasion pathways. Studies
combination of the ongoing exploration of the indi- on resolvins (64, 88, 188, 234) have shown that stimu-
vidual0 s own genome (68, 145, 147, 177–184, 193) and lating resolution pathways goes a long way to restor-
computer science (63) should make it possible to ing tissue homeostasis and periodontal healing.
generate specific microbiomes for each individual Addressing the underlying inflammatory imbalance
using metagenomic codes for identifying health and by harnessing the body’s own immunoregulatory
disease. These new approaches may change our mechanisms through the recruitment of regulatory
current concepts concerning the prevention, diagno- T-cells may reveal another possibility for the treat-
sis and treatment of periodontal diseases (239, 241). ment of periodontal disease. A recent study has
The concepts of keystone pathogens, along with shown that this can be accomplished by the sustained
polymicrobial synergy and dysbiosis, may serve as the release of small quantities of the chemokine CCL22
basis for more promising therapeutic options in peri- from a point source. Administration of these regula-
odontal disease. The health benefits of antibiotic use tory T-cell-recruiting treatments to the gingiva of
are undoubtedly a major success story of medicine mice and dogs reduced the clinical scores of peri-
but the strategy to target individual periopathogens, odontitis as well as hard- and soft-tissue destruction
such as P. gingivalis, is complicated because the (67). By enhancing protective innate immunity in
organism is difficult to eliminate completely. Besides ways that counteract chemokine paralysis, toll-like
having pathogenic effects, even at low abundance, receptor-4 antagonism (128) or human antimicrobial
P. gingivalis is able to survive inside epithelial cells, peptides (47) are considered to be very promising
well protected from antibiotics and thus is a source templates for the development of new clinical strate-
for recrudescence of infection (54, 107). Moreover, gies in immunology and cancer research without
overuse of these drugs has contributed to the emer- compromising the host.
gence of resistant microorganisms, which has become As an alternate strategy to antibiotic treatment,
a major public health problem (6, 14, 26). Studies manipulation of the microbiome through probiotics
have revealed a major reservoir of antibiotic-resis- and/or genetic modification of bacteria through syn-
tance genes in the human microbiome (62). There- thetic biology represent promising newer antimicro-
fore, some existing antibiotic treatments may lead to bial approaches. Probiotics containing Lactobacillus,
lethal treatment failures. If the use of antibiotics is Bifidobacterium and Saccharomyces are available for
the treatment of choice, then there is an urgent need treatment of traveler’s diarrhea and antibiotic-
to discover a new generation of antibiotics (123). induced diarrhea. Synthetic biology has already
As polymicrobial diseases in general are difficult to proved successful in applications for influenza vac-
treat with antibiotics, Guo et al. (82) suggested the cine production and makes it possible to develop
potential of using targeted antimicrobials to modu- modified species that can be introduced into the
late the microbiome. In a recent study of the cario- human body and monitored. Probiotic or engineered
genic Streptococcus mutans group, removal could be strains can be improved for efficiency, altering path-
achieved within an in vitro oral multispecies setting ways that may result in increased production of vita-
using a synthetic antimicrobial peptide: C16G2. In mins or the delivery of bioactive agents. These
addition, a drastic reconstruction of the multispecies modified species include yeasts, small eukaryotes or
communities was observed following treatment. bacteria that produce bioactive compounds at dis-
Another recent study, by Wu et al. (232), revealed the ease sites whilst also acting as a delivery system. They
identity of major genetic components involved in could also be used to produce synthetic natural-pro-
mediating the coaggregation of Candida albicans and duct medicines (222). In addition, synthetic virus bac-
F. nucleatum under planktonic conditions, whereas teriophages may be designed to modulate
mutation of FLO9 in C. albicans and radD in F. nu- populations of bacteria that have negative effects on
cleatum rendered the mutants defective in inter- human health (33). It is almost certain that there will
species binding. These observations of cellular further developments in microbial replacement ther-
components mediating co-adherence may be impor- apies and host-modulation strategies. However, as

103
Jepsen & Jepsen

Fig. 7. Magnitude of the periodontal benefits of using systemic antibiotic adjuncts to the treatment of periodontitis vs.
health risks to society and the world’s ecosystems. In periodontics, antibiotics should only be used as a last resort (Cour-
tesy: G. Armitage).

new therapies are introduced to restore a health- additional 0.4 mm in pocket depth reduction and
associated homeostasis between the periodontal 0.3 mm in clinical attachment level gain.
microbiota and the host, diagnostic tests will be In conclusion, the slight additional benefits of
required to assess treatment outcomes. adjunctive antimicrobials, which have been shown
for moderate forms of periodontitis, have to be bal-
anced against their side effects, and therefore their
Summary and conclusion prescription should be limited as much as possible.
Therapists should be careful not to underestimate the
A large number of randomized clinical trials and sys- effect of proper mechanical debridement and modifi-
tematic reviews have clearly established that adjunc- cation of behavioral risk factors. The patient’s overall
tive systemic antibiotics, combined with mechanical risk for periodontitis must be considered, and excep-
debridement, offer clinical improvements additional tions that may indicate antibiotic treatments include
to those obtained with scaling and root planing alone. cases of early-onset disease if the periodontal infec-
These effects are more pronounced in aggressive peri- tion needs to be rapidly suppressed, localized deep
odontitis compared with chronic periodontitis and in sites with persistent or recurrent disease and patients
initially deep pockets, whereas only limited additional with uncontrolled diabetes. However, these patients
improvements can be expected for moderately deep should ideally be treated in the practice setting of a
sites. The marginal clinical benefit of 0.3 mm of addi- specialist.
tional pocket reduction and 0.2 mm of additional
clinical attachment gain in patients with moderate
disease has to be balanced against possible severe References
risks (Fig. 7), such as microbiologically adverse effects
and increase in bacterial resistance. The effects of the 1. American Academy of Periodontology Task Force Report
adjunctive local administration of antimicrobials on the update to the 1999 Classification of Periodontal
Diseases and Conditions. J Periodontol 2015: 86: 835–838.
have also been very well documented in several sys-
2. Aimetti M, Romano F, Guzzi N, Carnevale G. Full-mouth
tematic reviews. Overall, in persistent or recurrent disinfection and systemic antimicrobial therapy in gener-
localized deep sites, antibiotic application by sus- alized aggressive periodontitis: a randomized, placebo-
tained local-delivery devices may offer a benefit of an controlled trial. J Clin Periodontol 2012: 39: 284–294.

104
Adjunctive antibiotic/antimicrobial therapy in periodontitis

3. Aimetti M, Romano F, Torta I, Cirillo D, Caposio P, 21. Boyton RJ, Reynolds CJ, Quigley KJ, Altmann DM. Immune
Romagnoli R. Debridement and local application of tetra- mechanisms and the impact of the disrupted lung micro-
cycline-loaded fibres in the management of persistent biome in chronic bacterial lung infection and bronchiecta-
periodontitis: results after 12 months. J Clin Periodontol sis. Clin Exp Immunol 2013: 171: 117–123.
2004: 31: 166–172. 22. Brusca SB, Abramson SB, Scher JU. Microbiome and
4. Akalin FA, Baltacioglu E, Sengu € n D, Hekimoglu S, Taskin mucosal inflammation as extra-articular triggers for
M, Etikan I, Fisenk I. A comparative evaluation of the clini- rheumatoid arthritis and autoimmunity. Curr Opin
cal effects of systemic and local doxycycline in the treat- Rheumatol 2014: 26: 101–107.
ment of chronic periodontitis. J Oral Sci 2004: 46: 25–35. 23. Cahenzli J, Ko € ller Y, Wyss M, Geuking MB, McCoy KD.
5. Al-Joburi W, Quee TC, Lautar C, Iugovaz I, Bourgouin J, Intestinal microbial diversity during early-life colonization
Delorme F, Chan ECS. Effects of adjunctive treatment of shapes long-term IgE levels. Cell Host Microbe 2013: 14:
periodontitis with tetracycline and spiramycin. J Periodon- 559–570.
tol 1989: 60: 533–539. 24. Cani PD. Gut microbiota and obesity: lessons from the
6. Aminov RI. The role of antibiotics and antibiotic resistance microbiome. Brief Funct Genomics 2013: 12: 381–387.
in nature. Environ Microbiol 2009: 11: 2970–2988. 25. Caricilli A, Saad M. The role of gut microbiota on insulin
7. Angaji M, Gelskey S, Nogueira-Filho G, Brothwell D. A sys- resistance. Nutrients 2013: 5: 829–851.
tematic review of clinical efficacy of adjunctive antibiotics 26. Carlet J. World alliance against antibiotic resistance: the
in the treatment of smokers with periodontitis. J Periodon- WAAAR declaration against antibiotic resistance. Med
tol 2010: 81: 1518–1528. Intensiva 2014: 39: 34–39; http://dx.doi.org/10.1016/
8. Antunes LCM, Han J, Ferreira RBR, Lolic P, Borchers CH, j.medin.2014.10.004.
Finlay BB. Effect of antibiotic treatment on the intestinal 27. Carvalho J, Novak MJ, Mota LF. Evaluation of the effect of
metabolome. Antimicrob Agents Chemother 2011: 55: subgingival placement of chlorhexidine chips as an
1494–1503. adjunct to scaling and root planing. J Periodontol 2007: 78:
9. Ardila CM, Granada MI, Guzma n IC. Antibiotic resistance 997–1001.
of subgingival species in chronic periodontitis patients. J 28. Casarin RC, Peloso Ribeiro ED, Sallum EA, Nocviti FH Jr,
Periodontal Res 2010: 45: 557–563. Goncßalves RB, Casati MZ. The combination of amoxicillin
10. Armitage GC. Development of a classification system for and metronidazole improves clinical and microbiologic
periodontal diseases and conditions. Ann Periodontol results of one-stage, full-mouth, ultrasonic debridement in
2000: 4: 1–6. aggressive periodontitis treatment. J Periodontol 2012: 83:
11. Armitage GC. Learned and unlearned concepts in peri- 988–998.
odontal diagnostics: a 50-year perspective. Periodontol 29. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife
2000 2013: 62: 20–36. WT, Schmidt TM, Young VB. Decreased diversity of the
12. Azmak DN, Atilla G, Luoto H, Sorsa T. The effect of subgin- fecal microbiome in recurrent Clostridium difficile-associ-
gival controlled-release delivery of chlorhexidine chip ated diarrhea. J Infect Dis 2008: 197: 435–438.
onclinical parameters and matrix metalloproteinase-8 levels 30. Chassaing B, Gewirtz AT. Gut microbiota, low-grade
in gingival crevicular fluid. J Periodontol 2002: 73: 603–615. inflammation, and metabolic syndrome. Toxicol Pathol
13. Baltacioglu E, Aslan M, Sarac O, Saybak A, Yuva P. Analysis 2014: 42: 49–53.
of clinical results of systemic antimicrobials combined 31. Cionca N, Giannopoulou C, Ugolotti G, Mombelli A.
with nonsurgical periodontal treatment for generalized Amoxicillin and metronidazole as an adjunct to full-mouth
aggressive periodontitis: a pilot study. J Can Dent Assoc scaling and root planing of chronic periodontitis. J Peri-
2011: 77: b97. odontol 2009: 80: 364–371.
14. Baquero F. Metagenomic epidemiology: a public health 32. Cionca N, Giannopoulou C, Ugolotti G, Mombelli A.
need for the control of antimicrobial resistance. Clin Microbiologic testing and outcomes of full-mouth scaling
Microbiol Infect 2012: 18: 67–73. and root planing with or without amoxicillin/metronida-
15. Becker W, Becker BE, Ochsenbein C, Kerry G, Caffesse R, zole in chronic periodontitis. J Periodontol 2010: 81: 15–23.
Morrison EC, Prichard J. A Longitudinal study comparing 33. Citorik RJ, Mimee M, Lu TK. Bacteriophage-based syn-
scaling, osseous surgery and modified Widman proce- thetic biology for the study of infectious diseases. Curr
dures. J Periodontol 1987: 59: 351–365. Opin Microbiol 2014: 19: 59–69.
16. Belda-Ferre P, Alcaraz LD, Cabrera-Rubio R, Romero H, 34. Cobb CM. Non-surgical pocket therapy. Ann Periodontol
Simo  n-Soro A, Pignatelli M, Mira A. The oral metagenome 1996: 1: 443–490.
in health and disease. ISME J 2011: 6: 46–56. 35. Cobb CM. Clinical significance of non-surgical periodontal
17. Bergstro € m J. Tobacco smoking and risk for periodontal therapy: an evidence-based perspective of scaling and root
disease. J Clin Periodontol 2003: 30: 107–113. planing. J Clin Periodontol 2002: 29(Suppl 2): 6–16.
18. Blaser M. Antibiotic overuse: stop the killing of beneficial 36. Costello EK, Stagaman K, Dethlefsen L, Bohannan B, Rel-
bacteria. Nature 2011: 476: 393–394. man DA. The application of ecological theory towards an
19. Bogren A, Teles RP, Torresyap G, Haffajee AD, Socransky understanding of the human microbiome. Science 2012:
SS, Wennstrom JL. Locally delivered doxycycline during 336: 1255–1262.
supportive periodontal therapy: a 3-year study. J Periodon- 37. Cosyn J, Wyn I, De Rouck T, Sabzevar MM. A chlorhexidine
tol 2008: 79: 827–835. varnish implemented treatment strategy for chronic peri-
20. Bonito DAJ, Lux L, Lohr KN. Impact of local adjuncts to odontitis. J Clin Periodontol 2005: 32: 750–756.
scaling and root planing in periodontal disease therapy: a 38. Cosyn J, Wyn I, De Rouck T, Sabzevar MM. Clinical bene-
systematic review. J Periodontol 2005: 76: 1227–1236. fits of subgingival chlorhexidine varnish application as an

105
Jepsen & Jepsen

adjunct to same-day full-mouth root planing: a pilot study. 55. Eickholz P, Kim T-S, Burklin T, Schacher B, Renggli HH,
J Periodontol 2006: 77: 1074–1079. Schaecken MT, Holle R, Kubler A, Ratka-Kru € ger P. Non-
39. Cosyn J, Wyn I, De Rouck T, Sabzevar MM. Subgingival surgical periodontal therapy with adjunctive topical doxy-
chlorhexidine varnish administration as an adjunct to cycline: a double-blind randomized controlled multicenter
same-day full-mouth root planing. I. clinical observations. study. (I). Study design and clinical results. J Clin Peri-
J Periodontol 2007: 78: 430–437. odontol 2002: 29: 108–117.
40. Cugini MA, Haffajee AD, Smith C, Kent RL, Socransky SS. 56. Ellen RP, McCulloch CAG. Evidence versus empiricism:
The effect of scaling and root planing on the clinical and rational use of systemic antimicrobial agents for
microbiological parameters of periodontal diseases: 12- treatment of periodontitis. Periodontol 2000 1996: 10:
month results. J Clin Periodontol 2000: 27: 30–36. 29–44.
41. Dannewitz B, Pohl S, Eickholz P, Kim TS. Clinical and 57. Feres M, Faveri M, Figueiredo LC, Teles R, Flemmig T, Wil-
microbiological effects of a combined mechanic-antibiotic liams R, Lang NP. Group B initiator paper non-surgical
therapy in subjects with Actinobacillus actinomycetem- periodontal therapy: mechanical debridement, antimicro-
comitans-associated periodontitis. Am J Dent 2007: 20: bial agents and other modalities. J Int Acad Periodontol
153–156. 2014: 17: s1–s10.
42. Dantas G, Sommer M, Oluwasegun RD, Church GM. 58. Feres M, Figueiredo LC, Silva Soares GM, Faveri M. Sys-
Bacteria subsisting on antibiotics. Science 2008: 320: 100– temic antibiotics in the treatment of periodontitis. Peri-
103. odontol 2000 2015: 67: 131–186.
43. Darby IB, Hodge PJ, Riggio MP, Kinane DF. Clinical and 59. Feres M, Soares GMS, Mendes JAV, Silva MP, Faveri M,
microbiological effect of scaling and root planing in smo- Teles R, Socransky SS, Figueiredo LC. Metronidazole alone
ker and non-smoker chronic and aggressive periodontitis or with amoxicillin as adjuncts to non-surgical treatment
patients. J Clin Periodontol 2005: 32: 200–206. of chronic periodontitis: a 1-year double-blinded, placebo-
44. D’Costa VM, King CE, Kalan L, Morar M, Sung WWL, Sch- controlled, randomized clinical trial. J Clin Periodontol
warz C, Froese D, Zazula G, Calmels F, Debruyne R, Gold- 2012: 39: 1149–1158.
ing GB, Poinar HN, Wright GD. Antibiotic resistance is 60. Finkelman RD, Polson AM. Evidence-based considerations
ancient. Nature 2011: 477: 457–461. for the clinical use of locally delivered, controlled-release
45. Delhaes L, Monchy S, Fre alle E, Hubans C, Salleron J, antimicrobials in periodontal therapy. J Dent Hyg 2013: 87:
Leroy S, Prevotat A, Wallet F, Wallaert B, Dei-Cas E, Sime- 249–264.
Ngando T, Chabe  M, Viscogliosi E. The airway microbiota 61. Flemmig TF, Milia n E, Karch H, Klaiber B. Differential clin-
in cystic fibrosis: a complex fungal and bacterial commu- ical treatment outcome after systemic metronidazole and
nity – implications for therapeutic management. PLoS amoxicillin in patients harboring Actinobacillus actino-
ONE 2012: 7: e36313. mycetemcomitans and/or Porphyromonas gingivalis. J Clin
46. Dethlefsen L, Relman DA. Colloquium paper: incomplete Periodontol 1998: 25: 380–387.
recovery and individualized responses of the human distal 62. Forslund K, Sunagawa S, Coelho LP, Bork P. Metagenomic
gut microbiota to repeated antibiotic perturbation. Proc insights into the human gut resistome and the forces that
Natl Acad Sci USA 2011: 108: 4554. shape it. BioEssays 2014: 36: 316–329.
47. Dommisch H, Jepsen S. Diverse functions of defensins and 63. Franzosa EA, Huang K, Meadow JF, Gevers D, Lemon KP,
other antimicrobial peptides in periodontal tissues. Peri- Bohannan B, Huttenhower C. Identifying personal
odontol 2000 2015: 69: 96–110. microbiomes using metagenomic codes. PNAS 2015:
48. Drisko CL. Nonsurgical periodontal therapy. Periodontol 112: E2930–8. www.pnas.org–cgi–doi–10.1073–pnas.14238
2000 2001: 25: 77–88. 54112.
49. Drisko CL. Periodontal debridement: still the treatment of 64. Freire MO, Van Dyke TE. Natural resolution of inflamma-
choice. J Evid Based Dent Pract 2014: 14: 33–41.e1. tion. Periodontol 2000 2013: 63: 149–164.
50. Dumas ME, Kinross J, Nicholson JK. Metabolic phenotyp- 65. Friesen LR, Williams KB, Krause LS, Killoy WJ. Controlled
ing and systems biology approaches to understanding local delivery of tetracycline with polymer strips in the
metabolic syndrome and fatty liver disease. Gastroenterol- treatment of periodontitis. J Periodontol 2002: 73: 13–19.
ogy 2014: 146: 46–62. 66. Garcia Canas P, Khouly I, Sanz J, Loomer PM. Effectiveness
51. DuPont AW, DuPont HL. The intestinal microbiota and of systemic antimicrobial therapy in combination with
chronic disorders of the gut. Nat Rev Gastroenterol Hepatol scaling and root planing in the treatmentof periodontitis. J
2011: 8: 523–531. Am Dent Assoc 2015: 146: 150–163.
52. Ehmke B, Beikler T, Haubitz I, Karch H, Flemmig TF. Mul- 67. Glowacki AJ, Yoshizawa S, Timmerman MF. Prevention of
tifactorial assessment of predictors for prevention of peri- inflammation-mediated bone loss in murine and canine
odontal disease progression. Clin Oral Investig 2003: 7: periodontal disease via recruitment of regulatory lympho-
217–221. cytes. Proc Natl Acad Sci USA 2013: 110: 18525–18530.
53. Ehmke DB, Moter A, Beikler T, Milian E, Flemmig TF. 68. Gonzaga-Jauregui C, Lupski JR, Gibbs RA. Human genome
Adjunctive antimicrobial therapy of periodontitis: long- sequencing in health and disease. Annu Rev Med 2012: 63:
term effects on disease progression and oral colonization. 35–61.
J Periodontol 2005: 76: 749–759. 69. Goodson JM. Antimicrobial strategies for treatment of
54. Eick S, Pfister W. Efficacy of antibiotics against periodon- periodontal diseases. Periodontol 2000 1994: 5: 142–168.
topathogenic bacteria within epithelial cells: an in vitro 70. Goodson JM, Gunsolley JC, Grossi SG, Bland PS, Otomo-
study. J Periodontol 2004: 75: 1327–1334. Corgel J, Doherty F, Comiskey J. Minocycline HCl micro-

106
Adjunctive antibiotic/antimicrobial therapy in periodontitis

spheres reduce red-complex bacteria in periodontal dis- ment of chronic periodontitis. J Contemp Dent Pract 2008:
ease therapy. J Periodontol 2007: 78: 1568–1579. 9: 25–32.
71. Goodson JM, Haffajee AD, Socransky SS, Kent R, Teles R, 84. Haffajee AD, Socransky SS, Gunsolley JC. Systemic anti-
Hasturk H, Bogren A, Van Dyke T, Wennstro € m J, Lindhe J. infective periodontal therapy. A systematic review. Ann
Control of periodontal infections: a randomized controlled Periodontol 2003: 8: 115–181.
trial I. The primary outcome attachment gain and pocket 85. Hajishengallis G. Periodontitis: from microbial immune
depth reduction at treated sites. J Clin Periodontol 2012: subversion to systemic inflammation. Nat Rev Immunol
39: 526–536. 2015: 15: 30–44.
72. Gordon JI, Dewey KG, Mills DA, RM F. The human gut 86. Hanes PJ, Purvis JP. Local anti-infective therapy: pharma-
microbiota and undernutrition. Sci Transl Med 2012: 4: 1– cological agents. A systematic review. Ann Periodontol
6. 2003: 8: 79–98.
73. Gracßa MA, Watts TL, Wilson RF, Palmer RM. A randomized 87. Harks I, Koch R, Eickholz P, Hoffmann T, Kim T-S, Kocher
controlled trial of a 2% minocycline gel as an adjunct to T, Meyle J, Kaner D, Schlagenhauf U, Doering S, Holtfreter
non-surgical periodontal treatment, using a design with B, Gravemeier M, Harmsen D, Ehmke B. Is progression of
multiple matching criteria. J Clin Periodontol 1997: 24: periodontitis relevantly influenced by systemic antibiotics?
249–253. A clinical randomized trial. J Clin Periodontol 2015: 42:
74. Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, 832–842.
Wagner MA, Bennett BJ, Li L, DiDonato JA, Lusis AJ, Hazen 88. Hasturk H, Abdallah R, Kantarci A, Nguyen D, Giordano N,
SL. Transmission of atherosclerosis susceptibility with gut Hamilton J, Van Dyke TE. Resolvin E1 (RvE1) attenuates
microbial transplantation. J Biol Chem 2015: 290: 5647– atherosclerotic plaque formation in diet and inflamma-
5660. tion-induced atherogenesis. Arterioscler Thromb Vasc Biol
75. Grice EA, Segre JA. The human microbiome: our second 2015: 4: 1–12.
genome. Annu Rev Genomics Hum Genet 2012: 13: 151– 89. Haubek D, Johansson A. Pathogenicity of the highly leuko-
170. toxic JP2 clone of Aggregatibacter actinomycetemcomitans
76. Griffiths GS, Ayob R, Guerrero A, Nibali L, Suvan J, Moles and its geographic dissemination and role in aggressive
DR, Tonetti MS. Amoxicillin and metronidazole as an periodontitis. J Oral Microbiol 2014: 6: 1.
adjunctive treatment in generalized aggressive periodonti- 90. Heasman PA, Heasman L, Stacey F, McCracken GI. Local
tis at initial therapy or re-treatment: a randomized con- delivery of chlorhexidine gluconate (PerioChip) in peri-
trolled clinical trial. J Clin Periodontol 2011: 38: 43–49. odontal maintenance patients. J Clin Periodontol 2001: 28:
77. Grisi DC, Salvador SL, Figueiredo LC, Souza SLS, Novaes 90–95.
AB, Grisi MFM. Effect of a controlled-release chlorhexi- 91. Heasman L, Stacey F, Preshaw PM, McCracken GI, Hep-
dine chip on clinical and microbiological parameters of burn S, Heasman PA. The effect of smoking on periodontal
periodontal syndrome. J Clin Periodontol 2002: 29: 875– treatment response: a review of clinical evidence. J Clin
881. Periodontol 2006: 33: 241–253.
78. Grossi SG, Goodson JM, Gunsolley JC, Otomo-Corgel J, 92. Heller D, Varela VM, Silva-Senem MXE, Torres MCB,
Bland PS, Doherty F, Comiskey J. Mechanical therapy with Feres-Filho EJ, Colombo APV. Impact of systemic antimi-
adjunctive minocycline microspheres reduces red-com- crobials combined with anti-infective mechanical debride-
plex bacteria in smokers. J Periodontol 2007: 78: 1741– ment on the microbiota of generalized aggressive
1750. periodontitis: a 6-month RCT. J Clin Periodontol 2011: 38:
79. Guerrero A, Echeverrıa JJ, Tonetti MS. Incomplete adher- 355–364.
ence to an adjunctive systemic antibiotic regimen 93. Henderson RJ, Boyens JV, Holborow DW, Pack ARC. Scal-
decreases clinical outcomes in generalized aggressive peri- ing and root-planing treatment with adjunctive subgingi-
odontitis patients: a pilot retrospective study. J Clin Peri- val minocycline. A clinical pilot study over six months, of
odontol 2007: 34: 897–902. sites adjacent to and remote from the antibiotic applica-
80. Guerrero A, Griffiths GS, Nibali L, Suvan J, Moles DR, Lau- tion. J Int Acad Periodontol 2002: 4: 77–87.
rell L, Tonetti MS. Adjunctive benefits of systemic amoxi- 94. Herrera D, Alonso B, Leo  n R, Rolda
n S, Sanz M. Antimicro-
cillin and metronidazole in non-surgical treatment of bial therapy in periodontitis: the use of systemic antimi-
generalized aggressive periodontitis: a randomized pla- crobials against the subgingival biofilm. J Clin Periodontol
cebo-controlled clinical trial. J Clin Periodontol 2005: 32: 2008: 35: 45–66.
1096–1107. 95. Herrera D, Matesanz P, Bascones-Martinez DA, Sanz M.
81. Gu€ ntsch A, Erler M, Preshaw PM, Sigusch BW, Klinger G, Local and systemic antimicrobial therapy in periodontics.
Glockmann E. Effect of smoking on crevicular polymor- J Evid Based Dent Pract 2012: S1: 50–60.
phonuclear neutrophil function in periodontally healthy 96. Herrera D, Sanz M, Jepsen S, Needleman I, Roldan S. A
subjects. J Periodontal Res 2006: 41: 184–188. systematic review on the effect of systemic antimicrobials
82. Guo L, McLean JS, Yang Y, Eckert R, Kaplan CW, Kyme P, as an adjunct to scaling and root planing in periodontitis
Sheikh O, Varnum B, Lux R, Shi W, He X. Precision-guided patients. J Clin Periodontol 2002: 29(Suppl 3): 136–159.
antimicrobial peptide as a targeted modulator of human 97. Hirsch R, Deng H, Laohachai MN. Azithromycin in peri-
microbial ecology. Proc Natl Acad Sci USA 2015: 112: 7569– odontal treatment: more than an antibiotic. J Periodontal
7574. Res 2011: 47: 137–148.
83. Gupta R, Pandit N, Aggarwal S, Verma A. Comparative 98. Ho€ glund-Aberg C, Kwamin F, Claesson R, Dahl en G,
evaluation of subgingivally delivered 10% doxycycline Johansson A, Haubek D. Progression of attachment loss is
hyclate and xanthan-based chlorhexidine gels in the treat- strongly associated with presence of the JP2 genotype of

107
Jepsen & Jepsen

Aggregatibacter actinomycetemcomitans: a prospective otics in patients with untreated chronic periodontitis: a


cohort study of a young adolescent population. J Clin Peri- systematic review and meta-analysis. J Periodontal Res
odontol 2014: 41: 232–241. 2014: 50: 294–314.
99. Huang YJ, Boushey HA. The Microbiome and asthma. Ann 114. Killoy WJ. The clinical significance of local chemothera-
Am Thorac Soc 2014: 11: S48–S51. pies. J Clin Periodontol 2002: 29(Suppl 2): 22–29.
100. Hung H-C, Douglass CW. Meta-analysis of the effect of 115. Kinane DF, Radvar M. The effect of smoking on mechani-
scaling and root planing, surgical treatment and antibiotic cal and antimicrobial periodontal therapy. J Periodontol
therapies on periodontal probing depth and attachment 1997: 68: 467–472.
loss. J Clin Periodontol 2002: 29: 975–986. 116. Kinane DF, Radvar M. A six-month comparison of three
101. Jarrold CR, Allaker RP, Young KA, Heath MR, Hardie JM, periodontal local antimicrobial therapies in persistent
Lynch E. Clinical and microbiological effects of topical periodontal pockets. J Periodontol 1999: 70: 1–7.
minocycline in the treatment of elderly patients with peri- 117. Kleiman SC, Carroll IM, Tarantino LM, Bulik CM. Gut feel-
odontitis. Br Dent J 1997: 183: 51–56. ings: a role for the intestinal microbiota in anorexia ner-
102. Jeffcoat MK, Bray KS, Ciancio SG, Dentino AR, Fine DH, vosa? Int J Eat Disord 2015: 48: 449–451.
Gordon JM, Gunsolley JC, Killoy WJ, Lowenguth RA, Mag- 118. Knapp CW, Dolfing J, Ehlert PAI, Graham DW. Evidence of
nusson NI, Offenbacher S, Palcanis KG, Proskin HM, increasing antibiotic resistance gene abundances in
Finkelman RD, Flashner M. Adjunctive use of a subgingi- archived soils since 1940. Environ Sci Technol 2010: 44:
val controlled-release chlorhexidine chip reduces probing 580–587.
depth and improves attachment level compared with scal- 119. Krajmalnik-Brown R, Ilhan ZE, Kang DW, DiBaise JK.
ing and root planing alone. J Periodontol 1998: 69: 989– Effects of gut microbes on nutrient absorption and energy
997. regulation. Nutr Clin Pract 2012: 27: 201–214.
103. Jeffcoat MK, Palcanis KG, Weatherford TW, Reese M, 120. Ley RE, Hamady M, Lozupone C, Turnbaugh PJ, Ramey
Geurs NC, Flashner M. Use of a biodegradable chlorhexi- RR, Bircher JS, Schlegel ML, Tucker TA, Schrenzel MD,
dine chip in the treatment of adult periodontitis: clinical Knight R, Gordon JI. Evolution of mammals and their gut
and radiographic findings. J Periodontol 2000: 71: 256– microbes. Science 2008: 320: 1647–1651.
262. 121. de Lima Oliveira AP, de Faveri M, Gursky LC, Mestnik MJ,
104. Jepsen S, Deschner J, Braun A, Schwarz F, Eberhard J. Cal- Feres M, Haffajee AD, Socransky SS, Teles RP. Effects of
culus removal and the prevention of its formation. Peri- periodontal therapy on GCF cytokines in generalized
odontol 2000 2011: 55: 167–188. aggressive periodontitis subjects. J Clin Periodontol 2012:
105. Jepsen K, Jaeger A, Jepsen S. Esthetic and functional reha- 39: 295–302.
bilitation of a severely compromised central incisor: an 122. Lindhe J, Westfelt E, Nyman S, Socransky SS, Heijl L, Brat-
interdisciplinary approach. Int J Periodontics Restorative thall G. Healing following surgical/non-surgical treatment
Dent 2015: 35: e35–e43. of periodontal disease. A clinical study. J Clin Periodontol
106. Jernberg C, Lofmark S, Edlund C, Jansson JK. Long-term 1982: 9: 115–128.
impacts of antibiotic exposure on the human intestinal 123. Ling LL, Schneider T, Peoples AJ, Spoering AL, Engels I,
microbiota. Microbiology 2010: 156: 3216–3223. Conlon BP, Mueller A, Scha €berle TF, Hughes DE,
107. Johnson JD, Chen R, Lenton PA, Zhang G, Hinrichs JE, Epstein S, Jones M, Lazarides L, Steadman VA, Cohen
Rudney JD. Persistence of extracrevicular bacterial reser- DR, Felix CR, Fetterman KA, Millett WP, Nitti AG, Zullo
voirs after treatment of aggressive periodontitis. J Peri- AM, Chen C, Lewis K. A new antibiotic kills pathogens
odontol 2008: 79: 2305–2312. without detectable resistance. Nature 2015: 517: 455–
108. Johnson GK, Guthmiller JM. The impact of cigarette smok- 459.
ing on periodontal disease and treatment. Periodontol 124. Loesche WJ, Giordani JR, Soehren S, Karciroti M. The
2000 2007: 44: 178–194. nonsurgical treatment of patients with periodontal dis-
109. Jones AA, Kornman KS, Newbold DA, Manwell MA. Clini- ease: results after five years. J Am Dent Assoc 2002: 133:
cal and microbiological effects of controlled-release locally 311–320.
delivered minocycline in periodontitis. J Periodontol 1994: 125. Loesche WJ, Giordano M, Hujuel PP, Schwarez J, Smith
65: 1058–1066. BA. Metronidazole in periodontitis: reduced need for sur-
110. Kasaj A, Chiriachide A, Willershausen B. The adjunctive gery. J Clin Periodontol 1992: 19: 103–112.
use of a controlled-release chlorhexidine chip following 126. Loesche WJ, Schmidt E, Smith BA, Timmerman MF.
treatment with a new ultrasonic device in supportive peri- Effects of metronidazole on periodontal treatment needs.
odontal therapy: a prospective, controlled clinical study. J Periodontol 1991: 62: 247–257.
Int J Dent Hyg 2007: 5: 225–231. 127. Machion L, Andia DC, Lecio G, Nociti FH, Casati MZ, Sal-
111. Kaur N, Chen C-C, Luther J, Kao JY. Intestinal dysbiosis in lum AW, Sallum EA. Locally delivered doxycycline as an
inflammatory bowel disease. Gut Microbes 2014: 2: 211– adjunctive therapy to scaling and root planing in the treat-
216. ment of smokers: a 2-year follow-up. J Periodontol 2006:
112. Keestra JAJ, Grosjean I, Coucke W, Quirynen M, Teughels 77: 606–613.
W. Non-surgical periodontal therapy with systemic antibi- 128. Mancek-Keber M, Frank-Bertoncelj M, Hafner-Bratkovic
otics in patients with untreated aggressive periodontitis: a I, Smole A, Zorko M, Pirher N, Hayer S, Kralj-Iglic V, Roz-
systematic review and meta-analysis. J Periodontal Res man B, Ilc N, Horvat S, Jerala R. Toll-like receptor 4
2014: 50: 689–706. senses oxidative stress mediated by the oxidation of
113. Keestra JAJ, Grosjean I, Coucke W, Quirynen M, Teughels phospholipids in extracellular vesicles. Sci Signal 2015: 8:
W. Non-surgical periodontal therapy with systemic antibi- ra60.

108
Adjunctive antibiotic/antimicrobial therapy in periodontitis

129. Marsh PD. Dental plaque: biological significance of a bio- masked, randomized clinical trial of sfficacy and safety. J
film and community life-style. J Clin Periodontol 2011: 32: Periodontol 2013: 84: 715–724.
7–15. 142. Morrison EC, Ramfjord SP, Hill RW. Short-term effects of
130. Marsh PD, Moter A, Devine DA. Dental plaque biofilms: initial, nonsurgical periodontal treatment (hygienic
communities, conflict and control. Periodontol 2000 2010: phase). J Clin Periodontol 1980: 7: 199–211.
55: 16–35. 143. Nakagawa T, Yamada S, Oosuka Y, Saito A, Hosaka Y, Ishi-
131. Mascarenhas P, Gapski R, Al-Shammari K, Hill R, Soehren kawa T, Okuda K. Clinical and microbiological study of
S, Fenno JC, Giannobile WV, Wang DH-L. Clinical local minocycline delivery (Periocline) following scaling
response of azithromycin as an adjunct to non-surgical and root planing in recurrent periodontal pockets. Bull
periodontal therapy in smokers. J Periodontol 2005: 76: Tokyo Dent Coll 1991: 32: 63–70.
426–436. 144. Newman MG, Kornman KS, Doherty FM. A 6-month mul-
132. Matarazzo F, Figueiredo LC, Cruz SEB, Faveri M, Feres ti-center evaluation of adjunctive tetracycline fiber ther-
M. Clinical and microbiological benefits of systemic apy used in conjunction with scaling and root planing in
metronidazole and amoxicillin in the treatment of maintenance patients: clinical results. J Periodontol 1994:
smokers with chronic periodontitis: a randomized pla- 65: 685–691.
cebo-controlled study. J Clin Periodontol 2008: 35: 885– 145. Nibali L, Henderson B, Sadiq ST, Donos N. Genetic dysbio-
896. sis: the role of microbial insults in chronic inflammatory
133. Matesanz-Pe rez P, Garcıa-Gargallo M, Figuero E, Bas- diseases. J Oral Microbiol 2014: 6: 22962.
cones-Martınez A, Sanz M, Herrera D. A systematic review 146. Nociti FH Jr, Casati MZ, Duarte PM. Current perspective
on the effects of local antimicrobials as adjuncts to subgin- of the impact of smoking on the progression and
gival debridement, compared with subgingival debride- treatment of periodontitis. Periodontol 2000 2015: 67: 187–
ment alone, in the treatment of chronic periodontitis. J 210.
Clin Periodontol 2013: 40: 227–241. 147. Olson MV. Human genetic individuality. Annu Rev Geno-
134. Matuliene G, Pjetursson BE, Salvi GE, Schmidlin K, Brag- mics Hum Genet 2012: 13: 1–27.
ger U, Zwahlen M, Lang NP. Influence of residual pockets 148. Oteo A, Herrera D, Figuero E, O’Connor A, Gonza lez I,
on progression of periodontitis and tooth loss: results after Sanz M. Azithromycin as an adjunct to scaling and root
11 years of maintenance. J Clin Periodontol 2008: 35: 685– planing in the treatment of Porphyromonas gingivalis-
695. associated periodontitis: a pilot study. J Clin Periodontol
135. Meinberg TA, Barnes CM, Dunning DG, Reinhardt RA. 2010: 37: 1005–1015.
Comparison of conventional periodontal maintenance 149. Palmer AC, Kishony R. Understanding, predicting and
versus scaling and root planing with subgingival minocy- manipulating the genotypic evolution of antibiotic resis-
cline. J Periodontol 2002: 73: 167–172. tance. Nat Rev 2013: 14: 243–248.
136. Mestnik MJ, Feres M, Figueiredo LC, Duarte PM, Lira EAG, 150. Palmer RM, Matthews JP, Wilson RF. Adjunctive systemic
Faveri M. Short-term benefits of the adjunctive use of and locally delivered metronidazole in the treatment of
metronidazole plus amoxicillin in the microbial profile periodontitis: a controlled clinical study. Br Dent J 1998:
and in the clinical parameters of subjects with generalized 184: 548–552.
aggressive periodontitis. J Clin Periodontol 2010: 37: 353– 151. Palmer RM, Matthews JP, Wilson RF. Non-surgical peri-
365. odontal treatment with and without adjunctive metron-
137. Million M, Lagier JC, Yahav D, Paul M. Gut bacterial idazole in smokers and non-smokers. J Clin Periodontol
microbiota and obesity. Clin Microbiol Infect 2013: 19: 1999: 26: 158–163.
305–313. 152. Paolantonio M, D’Angelo M, Grassi RF, Perinetti G, Pic-
138. Miranda TS, Feres M, Perez-Chaparro PJ, Faveri M, Fig- colomini R, Pizzo G, Annunziata M, D’Archivio D, D’Ercole
ueiredo LC, Tamashiro NS, Bastos MF, Duarte PM. S, Nardi G, Guida L. Clinical and microbiologic effects of
Metronidazole and amoxicillin as adjuncts to scaling and subgingival controlled-release delivery of chlorhexidine
root planing for the treatment of type 2 diabetic subjects chip in the treatment of periodontitis: a multicenter study.
with periodontitis: 1-year outcomes of a randomized pla- J Periodontol 2008: 79: 271–282.
cebo-controlled clinical trial. J Clin Periodontol 2014: 41: 153. Pflughoeft KJ, Versalovic J. Human microbiome in health
890–899. and disease. Annu Rev Pathol 2012: 7: 99–122.
139. Mombelli A. Antimicrobial advances in treating periodon- 154. Pihlstrom B. Commentary: Treatment of periodontitis: key
tal diseases. In: Kinane DF, Mombelli A, editors. Frontiers principles include removing subgingival bacterial deposits;
of oral biology: periodontal disease. Basel: Karger AG, 2012: providing a local environment and education to support
133–148. ISSN 1320-2433. good home care; providing regular professional mainte-
140. Mombelli A, Almaghlouth A, Cionca N, Courvoisier DS, nance. J Periodontol 2014: 85: 655–656.
Giannopoulou C. Differential benefits of amoxicillin– 155. Pihlstrom BL, McHugh RB, Oliphant TH, Ortiz-Campos C.
metronidazole in different phases of periodontal therapy Comparison of surgical and nonsurgical treatment of peri-
in a randomized controlled crossover clinical trial. J Peri- odontal disease. A review of current studies and additional
odontol 2015: 86: 367–375. results after 61/2 years. J Clin Periodontol 1983: 10: 524–
141. Mombelli A, Cionca N, Almaghlouth A, De caillet F, Cour- 541.
voisier DS, Giannopoulou C. Are there specific benefits of 156. Pihlstrom BL, Ortiz-Campos C, McHugh RB. A random-
amoxicillin plus metronidazole in Aggregatibacter actino- ized four-year study of periodontal therapy. J Periodontol
mycetemcomitans-associated periodontitis? Double- 1981: 52: 227–242.

109
Jepsen & Jepsen

157. Pozo-Rubio T, Olivares M, Nova E, De Palma G, Mujico JR, 173. Sakellari D, Dimitra S, Ioannidis I, Ioannis I, Antoniadou
Ferrer MD, Marcos A, Sanz Y. Immune development and M, Malama A, Slini T, Theodora S, Konstantinidis A,
intestinal microbiota in celiac disease. Clin Dev Immunol Antonis K. Clinical and microbiological effects of adjunc-
2012: 2012: 1–12. tive, locally delivered chlorhexidine on patients with
158. Preus HR, Gunleiksrud TM, Sandvik L, Gjermo P, Baelum chronic periodontitis. J Int Acad Periodontol 2010: 12: 20–
V. A randomized, double-masked clinical trial comparing 26.
four periodontitis treatment strategies: 1-year clinical 174. Sampaio E, Rocha M, Figueiredo LC, Faveri M, Duarte PM,
results. J Periodontol 2013: 84: 1075–1086. Gomes Lira EA, Feres M. Clinical and microbiological
159. Prosser JI, Bohannan BJM, Curtis TP, Ellis RJ, Firestone effects of azithromycin in the treatment of generalized
MK, Freckleton RP, Green JL, Green LE, Killham K, Lennon chronic periodontitis: a randomized placebo-controlled
JJ, Osborn AM, Solan M, van der Gast CJ, Young JPW. The clinical trial. J Clin Periodontol 2011: 38: 838–846.
role of ecological theory in microbial ecology. Nat Rev 175. Sanz I, Alonso B, Carasol M, Herrera D, Sanz M. Nonsurgi-
Microbiol 2007: 5: 384–392. cal treatment of periodontitis. J Evid Based Dent Pract
160. Quirynen M, Teughels W, van Steenberghe D. Microbial 2012: 12: 76–86.
shifts after subgingival debridement and formation of bac- 176. Sanz M, Teughels W. Innovations in non-surgical peri-
terial resistance when combined with local or systemic odontal therapy: consensus report of the Sixth European
antimicrobials. Oral Dis 2003: 9(Suppl 1): 30–37. Workshop on Periodontology. J Clin Periodontol 2008: 35:
161. Rabelo CC, Feres M, Goncßalves C, Figueiredo LC, Faveri 3–7.
M, Tu Y-K, Chambrone L. Systemic antibiotics in the treat- 177. Schaefer AS, Bochenek G, Jochens A, Ellinghaus D, Dom-
ment of aggressive periodontitis. A systematic review and misch H, Guzeldemir-Akcakanat E, Graetz C, Harks I,
a Bayesian Network meta-analysis. J Clin Periodontol Jockel-Schneider Y, Weinspach K, Meyle J, Eickholz P, Lin-
2015: 42: 647–657. den GJ, Cine N, Nohutcu R, Weiss E, Houri-Haddad Y,
162. Rams TE, Degener JE, van Winkelhoff AJ. Prevalence of b- Iraqi F, Folwaczny M, Noack B, Strauch K, Gieger C,
lactamase-producing bacteria in human periodontitis. J Waldenberger M, Peters A, Wijmenga C, Yilmaz E, Lieb W,
Periodontal Res 2013: 48: 493–499. Rosenstiel P, Doerfer C, Bruckmann C, Erdmann J, Ko €nig
163. Rams TE, Degener JE, van Winkelhoff AJ. Antibiotic resis- I, Jepsen S, Loos BG, Schreiber S. Genetic evidence for
tance in human chronic periodontitis microbiota. J Peri- plasminogen as a shared genetic risk factor of coronary
odontol 2014: 85: 160–169. artery disease and periodontitis. Circ Cardiovasc Genet
164. Rams TE, Feik D, Young V, Hammond BF, Slots J. Entero- 2015: 8: 159–167.
cocci in human periodontitis. Oral Microbiol Immunol 178. Schaefer AS, Bochenek G, Manke T, Nothnagel M, Graetz
1992: 7: 249–252. C, Thien A, Jockel-Schneider Y, Harks I, Staufenbiel I, Wij-
165. Ramseier CA, Suvan JE. Behaviour change counselling for menga C, Eberhard J, Guzeldemir-Akcakanat E, Cine N,
tobacco use cessation and promotion of healthy lifestyles: Folwaczny M, Noack B, Meyle J, Eickholz P, Trombelli L,
a systematic review. J Clin Periodontol 2015: 42(Suppl 16): Scapoli C, Nohutcu R, Bruckmann C, Doerfer C, Jepsen S,
S47–S58. Loos BG, Schreiber S. Validation of reported genetic risk
166. Ready D, Pratten J, Roberts AP, Bedi R, Mullany P, Wilson factors for periodontitis in a large-scale replication study. J
M. Potential role of Veillonella spp. as a reservoir of trans- Clin Periodontol 2013: 40: 563–572.
ferable tetracycline resistance in the oral cavity. Antimi- 179. Schaefer A, Jepsen S, Loos BG. Periodontal genetics: a dec-
crob Agents Chemother 2006: 50: 2866–2868. ade of genetic association studies mandates better study
167. Renvert S, Persson GR. A systematic review on the use of designs. J Clin Periodontol 2010: 38: 103–107.
residual probing depth, bleeding on probing and furcation 180. Schaefer AS, Richter GM, Dommisch H, Reinartz M, Noth-
status following initial periodontal therapy to predict fur- nagel M, Noack B, Laine ML, Folwaczny M, Groessner-
ther attachment and tooth loss. J Clin Periodontol 2002: 29 Schreiber B, Loos BG, Jepsen S, Schreiber S. CDKN2BAS is
(Suppl 3): 82–89. associated with periodontitis in different European popu-

168. Ribeiro EDP, Bittencourt S, Zanin ICJ, Bovi Ambrosano lations and is activated by bacterial infection. J Med Genet
GM, Sallum EA, Nociti FH, Goncßalves RB, Casati MZ. Full- 2010: 48: 38–47.
mouth ultrasonic debridement associated with amoxicillin 181. Schaefer AS, Richter GM, Groessner-Schreiber B, Noack B,
and metronidazole in the treatment of severe chronic peri- Nothnagel M, Mokhtari N-EE, Loos BG, Jepsen S, Schrei-
odontitis. J Periodontol 2009: 80: 1254–1264. ber S. Identification of a shared genetic susceptibility locus
169. Riep B, Purucker P, Bernimoulin JP. Repeated local for coronary heart disease and periodontitis. PLoS Genet
metronidazole-therapy as adjunct to scaling and root 2009: 5: e1000378.
planing in maintenance patients. J Clin Periodontol 1999: 182. Schaefer AS, Richter GM, Nothnagel M, Laine ML,
26: 710–715. Noack B, Glas J, Schrezenmeir J, Groessner-Schreiber
170. Roberts AP, Mullany P. Oral biofilms: a reservoir of trans- B, Jepsen S, Loos BG, Schreiber S. COX-2 is associated
ferable, bacterial, antimicrobial resistance. Expert Rev Anti with periodontitis in Europeans. J Dent Res 2010: 89:
Infect Ther 2010: 8: 1441–1450. 384–388.
171. Russell CD, Griffiths SJ, Baillie JK, Haas J. Stratified infec- 183. Schaefer AS, Richter GM, Nothnagel M, Laine ML, Ruhling
tion medicine: a call to arms. Lancet Infect Dis 2014: 14: A, Schafer C, Cordes N, Noack B, Folwaczny M, Glas J,
451. Dorfer C, Dommisch H, Groessner-Schreiber B, Jepsen S,
172. Ryder MI. The influence of smoking on host responses Loos BG, Schreiber S. A 30 UTR transition within DEFB1 is
in periodontal infections. Periodontol 2000 2007: 43: 267– associated with chronic and aggressive periodontitis.
277. Genes Immun 2010: 11: 45–54.

110
Adjunctive antibiotic/antimicrobial therapy in periodontitis

184. Schaefer AS, Richter GM, Nothnagel M, Manke T, Dom- in smokers and non-smokers with refractory periodontitis:
misch H, Jacobs G, Arlt A, Rosenstiel P, Noack B, Groess- a 5-year study. J Periodontol 1999: 70: 761–771.
ner-Schreiber B, Jepsen S, Loos BG, Schreiber S. A 199. Sommer MOA, Dantas G, Church GM. Functional charac-
genome-wide association study identifies GLT6D1 as a terization of the antibiotic resistance reservoir in the
susceptibility locus for periodontitis. Hum Mol Genet 2010: human microflora. Science 2009: 325: 1128–1131.
19: 553–562. 200. Song X, Sun J, Mikalsen T, Roberts AP, Sundsfjord A. Char-
185. Schmidt C. Mental health: thinking from the gut. Nature acterisation of the plasmidome within Enterococcus fae-
2015: 518: S12–S15. calis isolated from marginal periodontitis patients in
186. Sekirov I, Finlay BB. The role of the intestinal microbiota Norway. PLoS ONE 2013: 8: e62248.
in enteric infection. J Physiol 2009: 587: 4159–4167. 201. Soskolne WA, Chajek T, Flashner M, Landau I, Stabholtz A,
187. Sellitto M, Bai G, Serena G, Fricke WF, Sturgeon C, Gajer P, Kolatch B, Lerner EI. An in vivo study of the chlorhexidine
White JR, Koenig SSK, Sakamoto J, Boothe D, Gicquelais R, release profile of the PerioChipTM in the gingival crevicu-
Kryszak D, Puppa E, Catassi C, Ravel J, Fasano A. Proof of lar fluid, plasma and urine. J Clin Periodontol 1998: 25:
concept of microbiome-metabolome analysis and delayed 1017–1021.
gluten exposure on celiac disease autoimmunity in geneti- 202. Teughels W, Dhondt R, Dekeyser C, Quirynen M. Treat-
cally at-risk infants. PLoS ONE 2012: 7: e33387. ment of aggressive periodontitis. Periodontol 2000 2014:
188. Serhan CN, Chiang N, Van Dyke TE. Resolving inflamma- 65: 107–133.
tion: dual anti-inflammatory and pro-resolution lipid 203. Thomson WM, Broadbent JM, Welch D, Beck JD, Poulton
mediators. Nat Rev 2008: 8: 349–361. R. Cigarette smoking and periodontal disease among 32-
189. Serrano C, Torres N, Valdivieso C, Castan ~ o C, Barrera M, year-olds: a prospective study of a representative birth
Cabrales A. Antibiotic resistance of periodontal pathogens cohort. J Clin Periodontol 2007: 34: 828–834.
obtained from frequent antibiotic users. Acta Odontol 204. Thomson WM, Shearer DM, Broadbent JM, Foster Page
Latinoam 2009: 22: 99–104. LA, Poulton R. The natural history of periodontal attach-
190. Sgolastra F, Gatto R, Petrucci A, Monaco A. Effectiveness ment loss during the third and fourth decades of life. J Clin
of systemic amoxicillin/metronidazole as adjunctive ther- Periodontol 2013: 40: 672–680.
apy to scaling and root planing in the treatment of chronic 205. Timmerman MF, Van der Weijden GA, Van Steenbergen
periodontitis: a systematic review and meta-analysis. J TJ, Mantel MS, De Graaff J, Van der Velden U. Evaluation
Periodontol 2012: 83: 1257–1269. of the long-term efficacy and safety of locally-applied
191. Sgolastra F, Petrucci A, Gatto R, Monaco A. Effectiveness minocycline in adult periodontitis patients. J Clin Peri-
of systemic amoxicillin/metronidazole as an adjunctive odontol 1996: 23: 707–716.
therapy to full-mouth scaling and root planing in the 206. Tomasi C, Wennstro € m JL. Locally delivered doxycycline
treatment of aggressive periodontitis: a systematic review improves the healing following non-surgical periodontal
and meta-Analysis. J Periodontol 2012: 83: 731–743. therapy in smokers. J Clin Periodontol 2004: 31: 589–595.
192. Sharon I, Morowitz MJ, Thomas BC, Costello EK, Relman 207. Tomasi C, Wennstro € m JL. Locally delivered doxycycline as
DA, Banfield JF. Time series community genomics analysis an adjunct to mechanical debridement at retreatment of
reveals rapid shifts in bacterial species, strains, and phage periodontal pockets: outcome at furcation sites. J Peri-
during infant gut colonization. Genome Res 2013: 23: 111– odontol 2011: 82: 210–218.
120. 208. Tonetti MS, Cortellini P, Carnevale G, Cattabriga M, de
193. Shimizu S, Momozawa Y, Takahashi A, Nagasawa T, Ashi- Sanctis M, Pini Prato GP. A controlled multicenter study of
kawa K, Terada Y, Izumi Y, Kobayashi H, Tsuji M, Kubo adjunctive use of tetracycline periodontal fibers in
M, Furuichi Y. A genome-wide association study of peri- mandibular class II furcations with persistent bleeding. J
odontitis in a japanese population. J Dent Res 2015: 94: Clin Periodontol 1998: 25: 728–736.
555–561. 209. Tonetti MS, Lang NP, Cortellini P, Suvan JE, Eickholz P,
194. Silva MP, Feres M, Oliveira Sirotto TA, Silva Soares GM, Fourmousis I, Topoll H, Vangsted T, Wallkamm B, on
Velloso Mendes JA, Faveri M, Figueiredo LC. Clinical and behalf of the European Research Group on Periodontology
microbiological benefits of metronidazole alone or with (ERGOPerio). Effects of a single topical doxycycline
amoxicillin as adjuncts in the treatment of chronic peri- administration adjunctive to mechanical debridement in
odontitis: a randomized placebo-controlled clinical trial. J patients with persistent/recurrent periodontitis but
Clin Periodontol 2011: 38: 828–837. acceptable oral hygiene during supportive periodontal
195. Silva-Senem MXE, Heller D, Varela VM, Torres MCB, therapy. J Clin Periodontol 2012: 39: 475–482.
Feres-Filho EJ, Colombo APV. Clinical and microbiological 210. Toprak E, Veres A, Michel J-B, Chait R, Hartl DL, Kishony
effects of systemic antimicrobials combined to an anti- R. Evolutionary paths to antibiotic resistance under
infective mechanical debridement for the management of dynamically sustained drug selection. Nat Genet 2011: 44:
aggressive periodontitis: a 12-month randomized con- 101–105.
trolled trial. J Clin Periodontol 2013: 40: 242–251. 211. Vaarala O. Human intestinal microbiota and type 1 dia-
196. Siqueira JF, Fouad AF, Ro ^ cßas IN. Pyrosequencing as a tool betes. Curr Diab Rep 2013: 13: 601–607.
for better understanding of human microbiomes. J Oral 212. Van der Velden U, Varoufaki A, Hutter JW, Xu L, Timmer-
Microbiol 2012: 4: 10743. DOI: 10.3402/jom.v4i0.10743. man MF, van Winkelhoff AJ, Loos BG. Effect of smoking
197. Slots J. Systemic antibiotics in periodontics. J Periodontol and periodontal treatment on the subgingival microflora. J
2004: 75: 1553–1565. Clin Periodontol 2003: 30: 603–610.
198. So€ der DB, Nedlich U, Jin LJ. Longitudinal effect of non- 213. Van Dyke TE, Offenbacher S, Braswell L, Lessem J.
surgical treatment and systemic metronidazole for 1 week Enhancing the value of scaling and root-planing: Arestin

111
Jepsen & Jepsen

clinical trial results. J Int Acad Periodontol 2002: 4: 72– odontitis patients in the Netherlands and Spain. J Clin
76. Periodontol 2005: 32: 893–898.
214. Varela VM, Heller D, Silva-Senem MX, Torres MCMB, 229. van Winkelhoff AJ, Tijhof CJ, De Graaff J. Microbiological
Colombo APV, Feres-Filho EJ. Systemic antimicrobials and clinical results of metronidazole plus amoxicillin ther-
adjunctive to a repeated mechanical and antiseptic ther- apy in Actinobacillus actinomycetemcomitans-associated
apy for aggressive periodontitis: a 6-month randomized periodontitis. J Periodontol 1992: 63: 52–63.
controlled trial. J Periodontol 2011: 82: 1121–1130. 230. Wong MY, Lu CL, Liu CM, Hou LT, Chang WK. Clinical
215. Veloo ACM, Seme K, Raangs E, Rurenga P, Singadji Z, response of localized recurrent periodontitis treated with
Wekema-Mulder G, van Winkelhoff AJ. Antibiotic suscepti- scaling, root planing, and tetracycline fiber. J Formos Med
bility profiles of oral pathogens. Int J Antimicrob Agents Assoc 1998: 97: 490–497.
2012: 40: 450–454. 231. Wrighton KC, Thomas BC, Sharon I, Miller CS, Castelle CJ,
216. Villedieu A, Diaz-Torres ML, Hunt N, McNab R, Spratt DA, VerBerkmoes NC, Wilkins MJ, Hettich RL, Lipton MS, Wil-
Wilson M, Mullany P. Prevalence of tetracycline resistance liams KH, Long PE, Banfield JF. Fermentation, hydrogen,
genes in oral bacteria. Antimicrob Agents Chemother 2003: and sulfur metabolism in multiple uncultivated bacterial
47: 878–882. phyla. Science 2012: 337: 1661–1665.
217. Villedieu A, Diaz-Torres ML, Roberts AP, Hunt N, McNab 232. Wu T, Cen L, Kaplan C, Zhou X, Lux R, Shi W, He X. Cellu-
R, Spratt DA, Wilson M, Mullany P. Genetic basis of ery- lar components mediating coadherence of candida albi-
thromycin resistance in oral bacteria. Antimicrob Agents cans and Fusobacterium nucleatum. J Dent Res 2015: 94:
Chemother 2004: 48: 2298–2301. 1432–1438.
218. Walker WA. Initial intestinal colonization in the human 233. Xajigeorgiou C, Sakellari D, Slini T, Baka A, Konstantinidis
infant and immune homeostasis. Ann Nutr Metab 2013: A. Clinical and microbiological effects of different antimi-
63: 8–15. crobials on generalized aggressive periodontitis. J Clin
219. Walker CB, Gordon JM, Magnussen I, Clark WB. A role for Periodontol 2006: 33: 254–264.
antibiotics in the treatment of refractory periodontitis. J 234. Xu Z-Z, Zhang L, Liu T, Park JY, Berta T, Yang R, Serhan
Periodontol 1993: 64: 772–781. CN, Ji R-R. Resolvins RvE1 and RvD1 attenuate inflamma-
220. Walker C, Karpinia K. Rationale for use of antibiotics in tory pain via central and peripheral actions. Nat Med 2010:
periodontics. J Periodontol 2002: 73: 1188–1196. 16: 592–597.
221. Walker CB, Karpinia K, Baehni P. Chemotherapeutics: 235. Yalcin F, Demirel K, Onan U. Evaluation of adjunctive
antibiotics and other antimicrobials. Periodontol 2000 tetracycline fiber therapy with scaling and root planing:
2004: 36: 146–165. short-term clinical results. Periodontal Clin Investig 1999:
222. Walker MC, Thuronyi BW, Charkoudian LK, Lowry B, Kho- 21: 23–27.
sla C, Chang MCY. Expanding the fluorine chemistry of liv- 236. _
Yek EC, Cintan S, Topcuoglu N, Kulekci G, Issever H, Kan-
ing systems using engineered polyketide synthase tarci A. Efficacy of amoxicillin and metronidazole combi-
pathways. Science 2013: 341: 1089–1094. nation for the management of generalized aggressive
223. W allberg M, Cooke A. Immune mechanisms in type 1 dia- periodontitis. J Periodontol 2010: 81: 964–974.
betes. Trends Immunol 2013: 34: 583–591. 237. Yilmaz S, Kut B, Gursoy H, Eren-Kuru B, Noyan U,
224. Warburton PJ, Palmer RM, Munson MA, Wade WG. Kadir T. Er:YAG laser versus systemic metronidazole as
Demonstration of in vivo transfer of doxycycline resistance an adjunct to nonsurgical periodontal therapy: a clini-
mediated by a novel transposon. Int J Antimicrob Agents cal and microbiological study. Photomed Laser Surg
2007: 60: 973–980. 2012: 39: 526–536.
225. Wennstro € m JL, Newman HN, MacNeill SR, Killoy WJ, Grif- 238. Zandbergen D, Slot DE, Cobb CM, Van der Weijden FA.
fiths GS, Gillam DG, Krok L, Needleman IG, Weiss G, Gar- The clinical effect of scaling and root planing and the con-
rett S. Utilisation of locally delivered doxycycline in non- comitant administration of systemic amoxicillin and
surgical treatment of chronic periodontitis. J Clin Peri- metronidazole: a systematic review. J Periodontol 2013: 84:
odontol 2001: 28: 753–761. 332–351.
226. Winkel EG, van Winkelhoff AJ, Timmerman MF, Van der 239. Zarco MF, Vess TJ, Ginsburg GS. The oral microbiome in
Velden U, Van der Weijden GA. Amoxicillin plus metron- health and disease and the potential impact on personal-
idazole in the treatment of adult periodontitis patients. A ized dental medicine. Oral Dis 2012: 18: 109–120.
double-blind placebo-controlled study. J Clin Periodontol 240. Zhang X, Shen D, Fang Z, Jie Z, Qiu X, Zhang C, Chen Y, Ji
2001: 28: 296–305. L. Human gut microbiota changes reveal the progression
227. van Winkelhoff AJ, Bosch-Tijhof CJ, Winkel EG, van der of glucose intolerance. PLoS ONE 2013: 8: e71108.
Reijden WA. Smoking affects the subgingival microflora in 241. Zhou J, He Z, Yang Y, Deng Y, Tringe SG, Timmerman MF.
periodontitis. J Periodontol 2001: 72: 666–671. High-throughput metagenomic technologies for complex
228. van Winkelhoff AJ, Herrera D, Oteo A, Sanz M. Antimicro- microbial community analysis: open and closed formats.
bial profiles of periodontal pathogens isolated from peri- mBio 2015: 6: e02288–14.

112

You might also like