ArticlePDF Available

Outcome of hip and knee periprosthetic joint infections caused by pathogens resistant to biofilm-active antibiotics: results from a prospective cohort study

Authors:

Abstract and Figures

Background Periprosthetic joint infections (PJI) caused by pathogens, for which no biofilm-active antibiotics are available, are often referred to as difficult-to-treat (DTT). However, it is unclear whether the outcome of DTT PJI is worse than those of non-DTT PJI. We evaluated the outcome of DTT and non-DTT PJI in a prospective cohort treated with a two-stage exchange according to a standardized algorithm. Methods Patients with hip and knee PJI from 2013 to 2015 were prospectively included and followed up for ≥ 2 years. DTT PJI was defined as growth of microorganism(s) resistant to all available biofilm-active antibiotics. The Kaplan–Meier survival analysis was used to compare the probability of infection-free survival between DTT and non-DTT PJI and the 95% confidence interval (95% CI) was calculated. ResultsAmong 163 PJI, 30 (18.4%) were classified as DTT and 133 (81.6%) as non-DTT. At a mean follow-up of 33 months (range 24–48 months), the overall treatment success was 82.8%. The infection-free survival rate at 2 years was 80% (95% CI 61–90%) for DTT PJI and 84% (95% CI 76–89%) for non-DTT PJI (p = 0.61). The following mean values were longer in DTT PJI than in non-DTT PJI: hospital stay (45 vs. 28 days; p < 0.001), prosthesis-free interval (89 vs. 58 days; p < 0.001) and duration of antimicrobial treatment (151 vs. 117 days; p = 0.003). Conclusions The outcome of DTT and non-DTT PJI was similar (80–84%), however, at the cost of longer hospital stay, longer prosthesis-free interval and longer antimicrobial treatment. It remains unclear whether patients undergoing two-stage exchange with a long interval need biofilm-active antibiotics. Further studies need to evaluate the outcome in patients treated with biofilm-active antibiotics undergoing short vs. long interval.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Archives of Orthopaedic and Trauma Surgery
https://doi.org/10.1007/s00402-018-2886-0
ORTHOPAEDIC SURGERY
Outcome ofhip andknee periprosthetic joint infections caused
bypathogens resistant tobiofilm-active antibiotics: results
fromaprospective cohort study
DorukAkgün1· CarstenPerka1· AndrejTrampuz1· NoraRenz1
Received: 4 October 2017
© Springer-Verlag GmbH Germany, part of Springer Nature 2018
Abstract
Background Periprosthetic joint infections (PJI) caused by pathogens, for which no biofilm-active antibiotics are available,
are often referred to as difficult-to-treat (DTT). However, it is unclear whether the outcome of DTT PJI is worse than those of
non-DTT PJI. We evaluated the outcome of DTT and non-DTT PJI in a prospective cohort treated with a two-stage exchange
according to a standardized algorithm.
Methods Patients with hip and knee PJI from 2013 to 2015 were prospectively included and followed up for ≥ 2years.
DTT PJI was defined as growth of microorganism(s) resistant to all available biofilm-active antibiotics. The Kaplan–Meier
survival analysis was used to compare the probability of infection-free survival between DTT and non-DTT PJI and the 95%
confidence interval (95% CI) was calculated.
Results Among 163 PJI, 30 (18.4%) were classified as DTT and 133 (81.6%) as non-DTT. At a mean follow-up of 33months
(range 24–48months), the overall treatment success was 82.8%. The infection-free survival rate at 2years was 80% (95%
CI 61–90%) for DTT PJI and 84% (95% CI 76–89%) for non-DTT PJI (p = 0.61). The following mean values were longer in
DTT PJI than in non-DTT PJI: hospital stay (45 vs. 28 days; p < 0.001), prosthesis-free interval (89 vs. 58 days; p < 0.001)
and duration of antimicrobial treatment (151 vs. 117 days; p = 0.003).
Conclusions The outcome of DTT and non-DTT PJI was similar (80–84%), however, at the cost of longer hospital stay,
longer prosthesis-free interval and longer antimicrobial treatment. It remains unclear whether patients undergoing two-stage
exchange with a long interval need biofilm-active antibiotics. Further studies need to evaluate the outcome in patients treated
with biofilm-active antibiotics undergoing short vs. long interval.
Keywords Periprosthetic joint infection· Difficult-to-treat· Outcome· Biofilm-active antibiotics
Introduction
Periprosthetic joint infection (PJI) is a rare but severe com-
plication after joint arthroplasty [1]. The reported incidence
in hip and knee arthroplasty ranges from 0.3 to 2.2% after
primary and up to 5.9% after revision surgery [24]. Numer-
ous studies reported high cure rates of PJI (72–100%) using
a two-stage prosthesis exchange [512]. Although the main
surgical procedures are well described (i.e., prosthesis reten-
tion, one-stage or two-stage exchange), the type and duration
of antimicrobial treatment are still discussed controversially
and are often not mentioned in published reports.
While the role of microbial biofilms in the pathogenesis
of PJI is well studied [13, 14], the impact of the type of
pathogen and its antimicrobial susceptibility on the treat-
ment outcome is not well investigated. This topic is gaining
* Doruk Akgün
doruk.akguen@charite.de
Carsten Perka
carsten.perka@charite.de
Andrej Trampuz
andrej.trampuz@charite.de
Nora Renz
nora.renz@charite.de
1 Corporate member ofFreie Universität Berlin,
Humboldt-Universität zu Berlin, andBerlin Institute
ofHealth, Center forMusculoskeletal Surgery, Charité,
Universitätsmedizin Berlin, Charitéplatz 1, 10117Berlin,
Germany
Archives of Orthopaedic and Trauma Surgery
1 3
importance in the era of rising antimicrobial resistance, tak-
ing into account that only few antimicrobial agents exhibit
anti-biofilm activity such as rifampin against staphylococcal
biofilms [1520] and ciprofloxacin, against Gram-negative
biofilms [2123]. Several clinical studies demonstrated
higher cure rates of staphylococcal PJI with rifampin-
containing regimens compared to regimens containing no
biofilm-active antimicrobials, if the prosthesis was retained
[24, 25].
Therefore, PJI caused by pathogens, for which no bio-
film-active antibiotics are available, are often referred to
as difficult-to-treat (DTT), including rifampin-resistant
staphylococci, fluoroquinolone-resistant Gram-negative
bacteria, enterococci and fungi (mainly Candida spp.) [26].
PJI caused by enterococci and fungi are defined as DTT
independent of the pathogen susceptibility, as anti-biofilm
antimicrobials against these pathogens are lacking, as also
shown by high failure rates in PJI due to these microorgan-
isms [2729].
In the literature, two-stage exchange with a long interval
(> 6 weeks) is recommended in case of a DTT PJI but it is
unclear whether the unavailability of biofilm-active antibi-
otics adversely influences the treatment outcome compared
to non-DTT PJI if a two-stage exchange is used. In this pro-
spective cohort study, we compared the outcome of DTT and
non-DTT PJI managed with a two-stage exchange arthro-
plasty and standardized antimicrobial treatment regimen.
Methods
Study design
This retrospective analysis of prospectively collected data
was conducted in a tertiary healthcare center, providing
advanced specialty care to a population of about four mil-
lion inhabitants and including a specialized referral center
for septic surgery. A standardized diagnostic and therapeutic
algorithm was applied, including the surgical and antimi-
crobial treatment (see details below). The study protocol
was reviewed and approved by the institutional ethics com-
mittee and was done in accordance with the Declaration of
Helsinki.
Study population
Consecutive patients with hip and knee PJI treated with
a two-stage exchange at our institution from April 2013
through December 2015 were included. Patients with native
septic arthritis, with incomplete data sets and those with
follow-up less than 24 months were excluded.
Data collection
Cases were identified from the institutional patient-based PJI
database and data obtained by reviewing electronic medical
charts of consecutive patients with hip and knee PJI (see def-
inition below) and completing a case report form. At admis-
sion, comorbidities, the Charlson Comorbidity Index score
(CCI) [30], surgical history of the infected joint, clinical
manifestation, laboratory values such as serum C-reactive
protein (CRP) and blood leukocytes and presumed route of
infection (intraoperative vs. hematogenous) were recorded.
In addition, the following data were extracted: number of
revision surgeries between stages, length of hospital stay,
total duration of antimicrobial therapy, serum CRP value at
the time of reimplantation, microbiological and pathological
results of revisions and reimplantation.
Definition ofperiprosthetic joint infection
In this cohort, PJI was diagnosed according to the proposed
European Bone and Joint Infection Society (EBJIS) crite-
ria [31], since these criteria were used in several outcome
studies [32, 33] and are increasingly applied due to their
high sensitivity in low-grade infections. According to this
classification system, PJI was confirmed when at least one
of the following criteria was fulfilled: (1) macroscopic puru-
lence around the prosthesis; (2) presence of sinus tract; (3)
increased synovial fluid leukocyte count and differential
(> 2000 leukocytes/µl or > 70% granulocytes); (4) signifi-
cant microbial growth in synovial fluid, periprosthetic tissue
or sonication culture of retrieved prosthesis components; (5)
positive histopathology, defined as ≥ 23 granulocytes per
10 high-power field, corresponding to the type II or type III
classification of the periprosthetic membrane [34]. DTT PJI
was diagnosed when at least one of the causing microorgan-
isms was resistant to biofilm-active antibiotics, including
rifampin-resistant staphylococci, enterococci, fluoroqui-
nolone-resistant Gram-negative bacteria and fungi [26].
Sonication was performed for 1min at 40kHz (BactoSonic,
Bandelin electronic, Berlin, Germany), the resulting sonica-
tion fluid was plated onto aerobic and anaerobic sheep blood
agar plates and incubated for 14 days.
Standardized surgical andantimicrobial treatment
algorithm
Surgical treatment
During the first stage of the two-stage prosthesis exchange,
all implant parts, necrotic tissue, bone cement and other
foreign material were removed, followed by debridement
Archives of Orthopaedic and Trauma Surgery
1 3
of the surrounding tissue. In hip joints, resection arthro-
plasty without cement spacer was performed, according to
the institutional standard practice, whereas in knee joints an
antibiotic-loaded static cement spacer was inserted. Cement
spacers were loaded with 4g of antibiotics per 40g PMMA,
typically gentamicin in combination with either clindamycin
or vancomycin. The time of reimplantation was determined
by the following criteria: the local status was satisfactory
(no discharge from the incision site, surgical wound healed,
soft tissues not compromised) and absence of laboratory
signs of persisting infection (increased CRP without other
known reason). In DTT PJI, a longer interval (> 6 weeks)
was preferred [26]. In case of suspected persistent infec-
tion, an additional revision surgery with debridement was
performed (and spacer exchange, if present).
Antimicrobial treatment
Antibiotics were routinely administered intravenously for the
first 2 weeks, followed by oral antimicrobials until reimplan-
tation. If no appropriate oral antibiotics were available, an
outpatient intravenous antimicrobial therapy was adminis-
tered. Antibiotics were continued until reimplantation with-
out an antibiotic-free interval and without a pre-implantation
diagnostic joint aspiration. After reimplantation, antibiotics
were initially administered intravenously until the wound
was healed, followed by an oral biofilm-active (in non-DTT
PJI) or biofilm-non-active antimicrobials (in DTT PJI) for a
total treatment duration of at least 12 weeks, with a mini-
mum of 6 weeks antimicrobial course after reimplantation.
Evaluation offollow‑up
Patients were evaluated in our outpatient clinic 3, 6, 12
months after septic revision surgery and annually afterwards.
A standardized questionnaire evaluating the general health,
joint and skin status, any additional surgical interventions,
clinical complaints, and antibiotic use was used. Clinical,
laboratory and radiological evaluations were performed and
evaluated by an orthopedic surgeon and an infectious dis-
ease specialist. Further follow-up was performed contacting
the patients by phone or during the visit in the outpatient
clinic. Treatment outcome of PJI was evaluated accord-
ing to the modified Delphi international multidisciplinary
consensus criteria [35]. Infection-free outcome was defined
when all of the following criteria were fulfilled: (1) infec-
tion eradication, characterized by a healed wound without
sinus tract, drainage, and no recurrence of the infection at
last follow-up, (2) no subsequent surgical intervention after
reimplantation for persistent or perioperative infection, (3)
no PJI-related death, and (4) no long-term antimicrobial sup-
pression therapy (duration > 6 months) after reimplantation.
Due to poor sensitivity and specificity, laboratory parameters
such as CRP were not used for determination of infection
eradication [36].
Statistical analysis
Chi-square and Fisher’s exact tests were employed to com-
pare differences between categorical variables. The two-
sample t test (for parametric distribution) or Mann–Whitney
U test (for non-parametric distribution) was used to compare
continuous variables between groups. A multiple logistic
regression model in forward stepwise was used to determine
factors associated with DTT PJI evaluating the following
variables: (1) age-adjusted Charlson comorbidity index; (2)
age; (3) joint; (4) sex; (5) number of previous septic revision;
(6) sinus tract. The probability of infection-free survival and
the 95% confidence interval (95% CI) was estimated using
the Kaplan–Meier survival method. Survival curves between
groups were compared by the log-rank (Mantel–Cox) test. A
p value < 0.05 was considered significant. Statistical analysis
was performed using SPSS version 20 software (SPSS Inc.,
Chicago, IL, USA) and the software Prism (Version 7.03;
GraphPad, La Jolla, CA, USA).
Results
Patient demographics andinfection characteristics
Demographic data, clinical and laboratory findings of PJI
are summarized in Table1. During the study period, 182
patients undergoing two-stage exchange were identified.
After exclusion of 19 PJI due to a follow-up shorter than
24 months, 163 patients were included, 84 with hip and 79
with knee prostheses. Data of this patient cohort analyzing
the effect of positive microbiology at reimplantation on sub-
sequent treatment failure were recently published [37]. In 30
PJI (18 hip and 12 knee prostheses), PJI was categorized as
DTT, caused by enterococci in 18 patients, rifampin-resist-
ant Staphylococcus epidermidis in 10 patients and fungi in
3 patients. One patient had a polymicrobial infection with
Enterococcus faecalis and Candida albicans. No infection
caused by fluoroquinolone-resistant Gram-negative bacteria
was found. History of a previous septic revision was reported
in 70% (21 of 30) of patients in DTT group compared to 46%
(61 of 133) of patients in non-DTT group (p = 0.025). Mixed
infection was documented more frequently in DTT group
than non-DTT group (Table1).
Treatment
Table2 summarizes data on treatment characteristics. The
hospital stay (45 vs. 28 days, p < 0.001) and the prosthesis-
free interval (89 vs. 58 days; p < 0.001) were significantly
Archives of Orthopaedic and Trauma Surgery
1 3
longer in the DTT PJI group. The total duration of antimicro-
bial treatment was longer in the DTT PJI group than in the
non-DTT PJI group (151 vs. 117 days; p = 0.003).
Outcome analysis
At a mean follow-up of 33months (range 24–48months),
the overall treatment success was 82.8%. Figure1 shows the
Kaplan–Meier-estimated infection-free survival of DTT PJI
and non-DTT PJI group. The infection-free survival rate at
2years was 80% (95% CI 61–90%) for DTT PJI group and
84% for non-DTT PJI group (95% CI 76–89%) (p = 0.614).
Two of six failures in DTT PJI group were suppressed
with long-term antibiotics and the remaining four needed
a revision surgery due to a persistent infection, two cases
with isolation of the same microorganism and two cases with
other microorganisms. Two cases in the non-DTT PJI group
failed due to implemented long-term suppression antibiotic
therapy and 20 cases relapsed requiring another surgical
revision. None of the patients with suppression therapy (in
both groups) needed further surgical treatment.
Analyzing affected hip and knee prosthesis with differ-
ent surgical approach (i.e., use of cement spacers in knee
and resection arthroplasty in hip joints), the treatment suc-
cess was significantly better in hip than knee prosthesis. In
hip prosthesis cure of PJI was achieved in 75 of 84 patients
(89%) [including 15 of 18 patients (83%) in DTT group
and 60 of 66 patients (91%) for non-DTT], whereas in knee
Table 1 Patient demographics, clinical and laboratory findings
* The values are given as the mean and the standard deviation
a The values are given as the number with the percentage of the group in parentheses
Variable All patients (n = 163) DTT PJI group (n = 30) Non-DTT PJI group
(n = 133)
p value
Patient age at the time of explantation (years)* 70 ± 10 69.5 ± 12.5 69.6 ± 9.8 0.965
Sexa0.978
Male 71 (43.6) 13 (43.3) 58 (43.6)
Female 92 (56.4) 17 (56.7) 75 (56.4)
Jointa0.304
Hip 84 (51.5) 18 (60) 66 (49.6)
Knee 79 (48.5) 12 (40) 67 (50.4)
Charlson comorbidity index (age adjusted)*4 ± 2.1 4.5 ± 2.6 3.9 ± 2 0.122
CRP at admission (mg/l)*39.2 ± 67 41 ± 49.9 38.8 ± 70.5 0.053
Microbiologya
Monomicrobial 79 (48.5) 14 (46.7) 65 (48.9) 0.827
Polymicrobial 61 (37.4) 16 (53.3) 45 (33.8) 0.046
Negative cultures 23 (14.1) 23 (17.3)
No. of patients with previous septic revisionsa82 (50.3) 21 (70) 61 (45.9) 0.025
No. of previous septic revisions* 2.5 ± 1.6 1.8 ± 1.7 1.1 ± 1.7 0.015
Table 2 Treatment and outcome characteristics of all patients
The values are given as the mean and the standard deviation
a The four patients who were suppressed with long-term antimicrobial therapy are excluded
Variable All patients (n = 163) DTT PJI group (n = 30) Non-DTT PJI group
(n = 133)
p value
Treatment
Time until reimplantation (days) 63.9 ± 34.5 89.4 ± 50.5 58.1 ± 26.9 < 0.001
CRP prior to reimplantation (mg/l) 11.9 ± 13.1 10 ± 11.8 12.3 ± 13.4 0.164
Total duration of antimicrobial therapy (days) 123.3 ± 57.7 150.8 ± 74.7 117.4 ± 51.7 0.003
Duration of i.v. antimicrobial therapy (days)a32.8 ± 20.2 50.4 ± 31 29 ± 14.7 < 0.001
Duration of oral antimicrobial therapy (days)a84.1 ± 32.2 90.1 ± 47.3 82.8 ± 28.2 0.553
No. of revisions during interval 1.7 ± 1.3 2 ± 1.6 1.5 ± 1 0.324
Duration of hospital stay (days) 31.2 ± 15.9 44.5 ± 26.5 28.2 ± 10.4 < 0.001
Archives of Orthopaedic and Trauma Surgery
1 3
prosthesis PJI was cured in 59 of 79 patients (75%) [includ-
ing 8 of 12 patients (75%) in DTT group and 51 of 67 (76%)
in non-DTT group] (p = 0.023).
Figure2 shows survival curves of three groups with DTT
PJI caused by different microorganisms, including rifampin-
resistant staphylococci, enterococci and fungi. One patient
with polymicrobial infection involving Enterococcus faeca-
lis and Candida albicans was excluded from this sub-group
analysis. No significant differences were seen between the
DTT and non-DTT PJI sub-groups (p = 0.525).
Multiple regression analysis showed higher Charlson
comorbidity index as the only independent risk factor
predictive of DTT PJI (odds ratio 1.3; 95% confidence inter-
val 1.0–1.6; p = 0.046). The number of previous septic revi-
sions in history (1.8 vs. 1.1; p = 0.02) was also significantly
higher in DTT PJI group, but did not remain significant in
the multiple regression analysis (odds ratio 1.3; 95% CI
1.0–1.6; p = 0.059) (Table3).
Discussion
We report an overall treatment success of 82.8%, which is
rather high taking into consideration that the cohort con-
sisted predominantly of complex PJI cases, often referred
to our specialized septic surgery unit after several previous
treatment attempts have failed. Multiple regression analysis
did not show association of previous septic revisions with
DTT PJI. Nevertheless, the history of prior revisions for
infection, especially when combined with insufficient anti-
microbial therapy, is considered as one of the leading factors
associated with emergence of antimicrobial resistance and
consecutive treatment failure. In particular, rifampin mono-
therapy and repeated prior surgical revisions were associated
with emergence of rifampin resistance [38].
Many authors suggested that treating DTT PJI is more
challenging than in non-DTT PJI, requiring two-stage
exchange and a long prosthesis-free interval (≥ 6 weeks)
[26]. In our cohort, antibiotic-free period before reimplan-
tation was not used since discontinuation of antimicrobi-
als may allow recovery of microbial growth and thereby
increase the risk of treatment failure. In addition, joint
aspiration before reimplantation showed low sensitivity
and specificity for detection of persistent infection and was,
therefore, omitted [3941].
To our knowledge, this is the first study investigating
the outcome of PJI caused by microorganisms, for which
biofilm-active antibiotics are not available. In our cohort,
the treatment outcome of DTT and non-DTT PJI was similar
(80% and 84%, respectively); however, a longer prosthesis-
free interval, longer hospital stay and longer antimicrobial
treatment were needed to achieve the local and laboratory
01020304
05
0
0
20
40
60
80
100
Time (months)
Probability of infection-free survival (%)
p = 0.61
DTT
Non-DTT
Fig. 1 The probability of infection-free survival of difficult-to-treat
(DTT) and non-DTT PJI group
01020304050
0
20
40
60
80
100
Time (months)
Pr
obabilityofinfection-free survival(%)
p = 0.52
Rifampin resistent Staphylococcus epi.
Enterococcus spp.
Candida spp.
Fig. 2 The probability of infection-free survival of three difficult-to-
treat microorganisms (Candida spp., Enterococcus spp., rifampin-
resistant staphylococci). One patient with polymicrobial infection
including Candida albicans and Enterococcus faecalis was excluded
from the analysis
Table 3 Results of multivariate regression analysis as independent
risk factors predictive of DTT PJI
Variable OR (95% CI) p value
Age-adjusted CCI 1.3 (1.0–1.6) 0.046
Age 1.0 (0.9–1.0) 0.178
Joint 0.8 (0.5–1.2) 0.212
Sex 0.9 (0.6–1.4) 0.677
Number of previous septic
revisions
1.3 (1.0–1.6) 0.059
Sinus tract 1.0 (0.6–1.7) 0.998
Archives of Orthopaedic and Trauma Surgery
1 3
conditions required for reimplantation. This finding was
unexpected, since the unavailability of biofilm-active anti-
biotics was hypothesized to be associated with worse out-
come [42]. Based on findings of this study, the term DTT
PJI may not be appropriate, since the outcome is similar to
that in non-DTT PJI when the described surgical and antimi-
crobial treatment algorithm is followed. As recently stated,
a biofilm-active antibiotic should be used after reimplanta-
tion, if an implant (including spacer) is kept in place or the
new device is implanted before complete healing of infec-
tion [43]. As bacteria may also persist in the bone causing
chronic osteomyelitis, antibiotic treatment after reimplanta-
tion may be rational and may reduce the treatment failure
rate.
Antibiotic-impregnated cement spacers may act as a for-
eign body, on which biofilm may form despite initial high
local release of antibiotics [44]. Accordingly, a prosthesis-
free interval without the use of spacer is generally preferred
in difficult-to-treat infections [45]. However, our data are
demonstrating no differences between the DTT and non-
DTT group. Hence, based on these data, spacer may be used
in DTT PJI.
Several authors proposed other types of microorgan-
isms as causing DTT PJI, including methicillin-resistant
staphylococci, Pseudomonas aeruginosa and other resistant
microorganisms, but susceptible to biofilm-active antibiotics
[12, 4649]. However, the data supporting this conclusion
may represent a bias since most studies were retrospective,
without applying a standardized antimicrobial and surgical
treatment algorithm, including heterogeneous patients, not
reporting the type of antimicrobial treatment and using non-
uniform definitions of treatment failure [50]. In such inho-
mogeneous study population, the findings may be inconclu-
sive. This assumption is supported by several studies which
showed high cure rates (up to 92%) in methicillin-resistant
staphylococcal PJI, when rifampin was administered in com-
bination antimicrobial regimen, compared to low eradica-
tion rates when biofilm non-active agents were administered
alone, such as vancomycin, linezolid, daptomycin and fluo-
roquinolones [20, 51, 52]. Similarly, fluoroquinolones were
also active in the guinea pig foreign-body infection model in
terms of biofilm eradication in Gram-negative bacilli [26].
Therefore, use of biofilm-active antimicrobials and fol-
lowing a standardized therapeutic regime can achieve high
eradication rates in methicillin-resistant staphylococci, simi-
lar to methicillin-susceptible ones without prolonging the
prosthesis-free interval and total duration of antimicrobial
treatment.
We recognize few limitations of this study. First, the
low number of DTT PJI does not permit subgroup analy-
sis regarding the type of microorganism or best surgical
approach and the number of needed revisions. Therefore, it
is unknown whether the same treatment success in DTT PJI
would be achievable with less aggressive surgical regimen.
Second, the contribution of prolonged antimicrobial treat-
ment in DTT PJI on the high success is unknown since no
control group with shorter antimicrobial treatment course
was evaluated. Third, despite the minimum follow-up period
was 24 months, some patients may still develop an infection
relapse. Fourth, as in both groups, the prosthesis-free inter-
val and antibiotic treatment were longer than proposed by
the applied algorithm owing to the complexity of the mainly
secondarily referred patients, the interpretation of the results
is difficult. Fifth, unequal usage of spacers in hip and knee
patients can alter the treatment outcome and mislead our
outcome results.
Conclusion
The treatment outcome of DTT was similar to the one of
non-DTT PJI (80–84%), however, at the cost of longer hos-
pital stay, longer prosthesis-free interval and longer antimi-
crobial treatment in DTT PJI. It remains unclear whether
patients undergoing two-stage exchange with a long inter-
val need biofilm-active antibiotics. Further studies need to
evaluate the outcome in patients treated with biofilm-active
antibiotics undergoing short vs. long interval.
Compliance with ethical standards
Conflict of interest Authors DA, NR and AT declare that they have no
conflict of interest. Author CP reports grants from Aesculap, personal
fees from Zimmer, Smith & Nephew, Depuy/Synthes, Ceramtec and
Link, outside the submitted work.
References
1. Bozic KJ, Kurtz SM, Lau E, Ong K, Vail TP, Berry DJ (2009)
The epidemiology of revision total hip arthroplasty in the
United States. J Bone Joint Surg Am 91(1):128–133. https://doi.
org/10.2106/JBJS.H.00155
2. Kurtz SM, Ong KL, Lau E, Bozic KJ, Berry D, Parvizi J (2010)
Prosthetic joint infection risk after TKA in the medicare popula-
tion. Clin Orthop Relat Res 468(1):52–56. https://doi.org/10.1007/
s11999-009-1013-5
3. Ong KL, Kurtz SM, Lau E, Bozic KJ, Berry DJ, Parvizi J (2009)
Prosthetic joint infection risk after total hip arthroplasty in the
medicare population. J Arthroplasty 24(6 Suppl):105–109. https://
doi.org/10.1016/j.arth.2009.04.027
4. Bongartz T, Halligan CS, Osmon DR, Reinalda MS, Bamlet WR,
Crowson CS, Hanssen AD, Matteson EL (2008) Incidence and
risk factors of prosthetic joint infection after total hip or knee
replacement in patients with rheumatoid arthritis. Arthritis Rheum
59(12):1713–1720. https://doi.org/10.1002/art.24060
5. De Man FH, Sendi P, Zimmerli W, Maurer TB, Ochsner PE,
Ilchmann T (2011) Infectiological, functional, and radiographic
outcome after revision for prosthetic hip infection according to a
strict algorithm. Acta Orthop 82(1):27–34. https://doi.org/10.31
09/17453674.2010.548025
Archives of Orthopaedic and Trauma Surgery
1 3
6. Engesaeter LB, Dale H, Schrama JC, Hallan G, Lie SA (2011)
Surgical procedures in the treatment of 784 infected THAs
reported to the Norwegian Arthroplasty Register. Acta Orthop
82(5):530–537. https://doi.org/10.3109/17453674.2011.62357
2
7. Haddad FS, Sukeik M, Alazzawi S (2015) Is single-stage revi-
sion according to a strict protocol effective in treatment of
chronic knee arthroplasty infections? Clin Orthop Relat Res
473(1):8–14. https://doi.org/10.1007/s11999-014-3721-8
8. Oussedik SI, Dodd MB, Haddad FS (2010) Outcomes of revi-
sion total hip replacement for infection after grading according
to a standard protocol. J Bone Joint Surg Br 92(9):1222–1226.
https://doi.org/10.1302/0301-620X.92B9.23663
9. Macheras GA, Kateros K, Galanakos SP, Koutsostathis SD,
Kontou E, Papadakis SA (2011) The long-term results of a two-
stage protocol for revision of an infected total knee replace-
ment. J Bone Joint Surg Br 93(11):1487–1492. https://doi.
org/10.1302/0301-620X.93B11.27319
10. Gooding CR, Masri BA, Duncan CP, Greidanus NV, Garbuz
DS (2011) Durable infection control and function with the
PROSTALAC spacer in two-stage revision for infected knee
arthroplasty. Clin Orthop Relat Res 469(4):985–993. https://
doi.org/10.1007/s11999-010-1579-y
11. Mortazavi SM, Vegari D, Ho A, Zmistowski B, Parvizi
J (2011) Two-stage exchange arthroplasty for infected
total knee arthroplasty: predictors of failure. Clin Orthop
Relat Res 469(11):3049–3054. https://doi.org/10.1007/
s11999-011-2030-8
12. Kurd MF, Ghanem E, Steinbrecher J, Parvizi J (2010) Two-stage
exchange knee arthroplasty: does resistance of the infecting organ-
ism influence the outcome? Clin Orthop Relat Res 468(8):2060–
2066. https://doi.org/10.1007/s11999-010-1296-6
13. Tzeng A, Tzeng TH, Vasdev S, Korth K, Healey T, Parvizi J,
Saleh KJ (2015) Treating periprosthetic joint infections as
biofilms: key diagnosis and management strategies. Diagn
Microbiol Infect Dis 81(3):192–200. https://doi.org/10.1016/j.
diagmicrobio.2014.08.018
14. McConoughey SJ, Howlin R, Granger JF, Manring MM, Calhoun
JH, Shirtliff M, Kathju S, Stoodley P (2014) Biofilms in peripros-
thetic orthopedic infections. Future Microbiol 9(8):987–1007.
https://doi.org/10.2217/fmb.14.64
15. Zimmerli W, Widmer AF, Blatter M, Frei R, Ochsner PE (1998)
Role of rifampin for treatment of orthopedic implant-related
staphylococcal infections: a randomized controlled trial. Foreign-
body infection (FBI) Study Group. JAMA 279(19):1537–1541
16. Widmer AF, Gaechter A, Ochsner PE, Zimmerli W (1992) Anti-
microbial treatment of orthopedic implant-related infections with
rifampin combinations. Clin Infect Dis 14(6):1251–1253
17. Drancourt M, Stein A, Argenson JN, Zannier A, Curvale G,
Raoult D (1993) Oral rifampin plus ofloxacin for treatment of
Staphylococcus-infected orthopedic implants. Antimicrob Agents
Chemother 37(6):1214–1218
18. Isiklar ZU, Darouiche RO, Landon GC, Beck T (1996) Efficacy
of antibiotics alone for orthopaedic device related infections. Clin
Orthop Relat Res (332):184–189
19. Chuard C, Herrmann M, Vaudaux P, Waldvogel FA, Lew DP
(1991) Successful therapy of experimental chronic foreign-body
infection due to methicillin-resistant Staphylococcus aureus
by antimicrobial combinations. Antimicrob Agents Chemother
35(12):2611–2616
20. John AK, Baldoni D, Haschke M, Rentsch K, Schaerli P, Zim-
merli W, Trampuz A (2009) Efficacy of daptomycin in implant-
associated infection due to methicillin-resistant Staphylococcus
aureus: importance of combination with rifampin. Antimicrob
Agents Chemother 53(7):2719–2724. https://doi.org/10.1128/
AAC.00047-09
21. Owusu-Ababio G, Rogers J, Anwar H (1999) Effectiveness of cip-
rofloxacin microspheres in eradicating bacterial biofilm. J Control
Release 57(2):151–159
22. Elkhatib W, Noreddin A (2014) Efficacy of ciprofloxacin-
clarithromycin combination against drug-resistant Pseudomonas
aeruginosa mature biofilm using invitro experimental model.
Microb Drug Resist 20(6):575–582. https://doi.org/10.1089/
mdr.2014.0024
23. Reffuveille F, Fuente-Nunez Cde L, Fairfull-Smith KE, Hancock
RE (2015) Potentiation of ciprofloxacin action against Gram-neg-
ative bacterial biofilms by a nitroxide. Pathog Dis 73 (5). https://
doi.org/10.1093/femspd/ftv016
24. El Helou OC, Berbari EF, Lahr BD, Eckel-Passow JE, Razonable
RR, Sia IG, Virk A, Walker RC, Steckelberg JM, Wilson WR,
Hanssen AD, Osmon DR (2010) Efficacy and safety of rifampin
containing regimen for staphylococcal prosthetic joint infections
treated with debridement and retention. European journal of clini-
cal microbiology & infectious diseases: official publication of
the European Society. of Clinical Microbiology 29(8):961–967.
https://doi.org/10.1007/s10096-010-0952-9
25. Giulieri SG, Graber P, Ochsner PE, Zimmerli W (2004) Manage-
ment of infection associated with total hip arthroplasty according
to a treatment algorithm. Infection 32(4):222–228. https://doi.
org/10.1007/s15010-004-4020-1
26. Zimmerli W, Moser C (2012) Pathogenesis and treat-
ment concepts of orthopaedic biofilm infections. FEMS
Immunol Med Microbiol 65(2):158–168. https://doi.
org/10.1111/j.1574-695X.2012.00938.x
27. Rasouli MR, Tripathi MS, Kenyon R, Wetters N, Della Valle
CJ, Parvizi J (2012) Low rate of infection control in entero-
coccal periprosthetic joint infections. Clin Orthop Relat Res
470(10):2708–2716. https://doi.org/10.1007/s11999-012-2374-8
28. Kheir MM, Tan TL, Higuera C, George J, Della Valle CJ, Shen
M, Parvizi J (2016) Periprosthetic Joint Infections Caused by
Enterococci Have Poor Outcomes. J Arthroplasty. https://doi.
org/10.1016/j.arth.2016.09.017
29. Ueng SW, Lee CY, Hu CC, Hsieh PH, Chang Y (2013) What is
the success of treatment of hip and knee candidal periprosthetic
joint infection? Clin Orthop Relat Res 471(9):3002–3009. https://
doi.org/10.1007/s11999-013-3007-6
30. Charlson ME, Pompei P, Ales KL, MacKenzie CR (1987) A new
method of classifying prognostic comorbidity in longitudinal stud-
ies: development and validation. J Chronic Dis 40(5):373–383
31. Ochsner PEBO., Bodler PM, Broger I, Eich G, Hefti F, Maurer T,
Nötzli H, Seiler S, Suva D, Trampuz A, Uckay I, Vogt M, Zim-
merli W (2016) Infections of the musculoskeletal system. Basic
principles, prevention, diagnosis and treatment. Swiss orthopae-
dics in-house publisher, Grandvaux
32. Akgun D, Trampuz A, Perka C, Renz N (2017) High failure
rates in treatment of streptococcal periprosthetic joint infection:
results from a seven-year retrospective cohort study. Bone Joint J
99-B(5):653–659. https://doi.org/10.1302/0301-620X.99B5.BJJ-
2016-0851.R1
33. Perez-Prieto D, Portillo ME, Puig-Verdie L, Alier A, Martinez
S, Sorli L, Horcajada JP, Monllau JC (2017) C-reactive protein
may misdiagnose prosthetic joint infections, particularly chronic
and low-grade infections. Int Orthop. https://doi.org/10.1007/
s00264-017-3430-5
34. Krenn V, Morawietz L, Perino G, Kienapfel H, Ascherl R, Has-
senpflug GJ, Thomsen M, Thomas P, Huber M, Kendoff D, Baum-
hoer D, Krukemeyer MG, Natu S, Boettner F, Zustin J, Kolbel
B, Ruther W, Kretzer JP, Tiemann A, Trampuz A, Frommelt L,
Tichilow R, Soder S, Muller S, Parvizi J, Illgner U, Gehrke T
(2014) Revised histopathological consensus classification of joint
implant related pathology. Pathol Res Pract 210(12):779–786.
https://doi.org/10.1016/j.prp.2014.09.017
Archives of Orthopaedic and Trauma Surgery
1 3
35. Diaz-Ledezma C, Higuera CA, Parvizi J (2013) Success after
treatment of periprosthetic joint infection: a Delphi-based inter-
national multidisciplinary consensus. Clin Orthop Relat Res
471(7):2374–2382. https://doi.org/10.1007/s11999-013-2866-1
36. Kusuma SK, Ward J, Jacofsky M, Sporer SM, Della Valle CJ
(2011) What is the role of serological testing between stages of
two-stage reconstruction of the infected prosthetic knee? Clin
Orthop Relat Res 469(4):1002–1008. https://doi.org/10.1007/
s11999-010-1619-7
37. Akgun D, Muller M, Perka C, Winkler T (2017) A positive bac-
terial culture during re-implantation is associated with a poor
outcome in two-stage exchange arthroplasty for deep infection.
Bone Joint J 99-B(11):1490–1495. https://doi.org/10.1302/0301-
620X.99B11.BJJ-2017-0243-R1
38. Achermann Y, Eigenmann K, Ledergerber B, Derksen L, Rafeiner
P, Clauss M, Nuesch R, Zellweger C, Vogt M, Zimmerli W
(2013) Factors associated with rifampin resistance in staphy-
lococcal periprosthetic joint infections (PJI): a matched case-
control study. Infection 41(2):431–437. https://doi.org/10.1007/
s15010-012-0325-7
39. Hoell S, Moeller A, Gosheger G, Hardes J, Dieckmann R, Schulz
D (2016) Two-stage revision arthroplasty for periprosthetic joint
infections: what is the value of cultures and white cell count in
synovial fluid and CRP in serum before second stage reimplan-
tation? Arch Orthop Trauma Surg 136(4):447–452. https://doi.
org/10.1007/s00402-015-2404-6
40. Osmon DR (2016) Microbiology and antimicrobial challenges
of prosthetic joint infection. J Am Acad Orthop Surg. https://doi.
org/10.5435/JAAOS-D-16-00639
41. Preininger B, Janz V, von Roth P, Trampuz A, Perka CF,
Pfitzner T (2017) Inadequacy of joint aspiration for detec-
tion of persistent periprosthetic infection during two-stage
septic revision knee surgery. Orthopedics. https://doi.
org/10.3928/01477447-20170411-04
42. Nana A, Nelson SB, McLaren A, Chen AF (2016) What’s new in
musculoskeletal infection: update on biofilms. J Bone Joint Surg
Am 98(14):1226–1234. https://doi.org/10.2106/JBJS.16.00300
43. Zimmerli W, Sendi P (2017) Orthopaedic biofilm infections.
APMIS 125(4):353–364. https://doi.org/10.1111/apm.12687
44. Nelson CL, Jones RB, Wingert NC, Foltzer M, Bowen TR
(2014) Sonication of antibiotic spacers predicts failure during
two-stage revision for prosthetic knee and hip infections. Clin
Orthop Relat Res 472(7):2208–2214. https://doi.org/10.1007/
s11999-014-3571-4
45. Zimmerli W, Trampuz A, Ochsner PE (2004) Prosthetic-joint
infections. N Engl J Med 351(16):1645–1654. https://doi.
org/10.1056/NEJMra040181
46. Zeller V, Lhotellier L, Marmor S, Leclerc P, Krain A, Graff W,
Ducroquet F, Biau D, Leonard P, Desplaces N, Mamoudy P (2014)
One-stage exchange arthroplasty for chronic periprosthetic hip
infection: results of a large prospective cohort study. J Bone Joint
Surg Am 96(1):e1. https://doi.org/10.2106/JBJS.L.01451
47. Trampuz A, Zimmerli W (2008) Diagnosis and treatment of
implant-associated septic arthritis and osteomyelitis. Curr Infect
Dis Rep 10(5):394–403
48. Parvizi J, Azzam K, Ghanem E, Austin MS, Rothman RH (2009)
Periprosthetic infection due to resistant staphylococci: serious
problems on the horizon. Clin Orthop Relat Res 467(7):1732–
1739. https://doi.org/10.1007/s11999-009-0857-z
49. Parry MC, Duncan CP (2014) The challenge of methicillin resist-
ant staphylococcal infection after total hip replacement: over-
looked or overstated? Bone Joint J 96-B(11 Supple A):60–65.
https://doi.org/10.1302/0301-620X.96B11.34333
50. Sendi P, Zimmerli W (2011) Challenges in periprosthetic knee-
joint infection. Int J Artif Organs 34(9):947–956. https://doi.
org/10.5301/ijao.5000032
51. Baldoni D, Haschke M, Rajacic Z, Zimmerli W, Trampuz A
(2009) Linezolid alone or combined with rifampin against methi-
cillin-resistant Staphylococcus aureus in experimental foreign-
body infection. Antimicrob Agents Chemother 53(3):1142–1148.
https://doi.org/10.1128/AAC.00775-08
52. Trampuz A, Murphy CK, Rothstein DM, Widmer AF, Landmann
R, Zimmerli W (2007) Efficacy of a novel rifamycin derivative,
ABI-0043, against Staphylococcus aureus in an experimental
model of foreign-body infection. Antimicrob Agents Chemother
51(7):2540–2545. https://doi.org/10.1128/AAC.00120-07
... Questions/purposes (1) In patients with hip and knee arthroplasties on the same side who experience a PJI of one implant, are there factors associated with the development of subsequent PJI of the other implant? (2) In this patient group, how often is the same organism responsible for both PJIs? (3) Is a shorter distance from an infected prosthetic joint to an ipsilateral prosthetic joint associated with greater odds of subsequent infection of the second joint? Methods We designed a retrospective study of a longitudinally maintained institutional database that identified all one-stage and two-stage procedures performed for chronic PJI of the hip and knee at our tertiary referral arthroplasty center between January 2010 and December 2018 (n = 2352). ...
... We therefore asked, (1) In patients with hip and knee arthroplasties on the same side who experience a PJI of one implant, are there factors associated with the development of subsequent PJI of the other implant? (2) In this patient group, how often is the same organism responsible for both PJIs? (3) Is a shorter distance from an infected prosthetic joint to an ipsilateral prosthetic joint associated with greater odds of subsequent infection of the second joint? ...
... Age in years, mean 6 SD 77 6 9.6 BMI in kg/m 2 , mean 6 SD 30 6 6. differ in terms of bacterial virulence (high and low virulence were defined by Morgenstern et al. [19]), polymicrobial infection, and difficult-to-treat germs (defined as those caused by rifampicin-resistant staphylococci, fluoroquinoloneresistant streptococci, enterococci, and fungi [2,7]). Of the patients, 65% (64 of 98) were female; the mean patient age was 77 6 9.6 years, and the mean BMI was 30 6 6.2 kg/m 2 . ...
Article
Background: Periprosthetic joint infection (PJI), the most common cause of revision after TKA and THA, is a devastating complication for patients that is difficult to diagnose and treat. An increase in the number of patients with multiple joint arthroplasties in the same extremity will result in an increased risk of ipsilateral PJI. However, there is no definition of risk factors, micro-organism patterns, and safe distance between knee and hip implants for this patient group. Questions/purposes: (1) In patients with hip and knee arthroplasties on the same side who experience a PJI of one implant, are there factors associated with the development of subsequent PJI of the other implant? (2) In this patient group, how often is the same organism responsible for both PJIs? (3) Is a shorter distance from an infected prosthetic joint to an ipsilateral prosthetic joint associated with greater odds of subsequent infection of the second joint? Methods: We designed a retrospective study of a longitudinally maintained institutional database that identified all one-stage and two-stage procedures performed for chronic PJI of the hip and knee at our tertiary referral arthroplasty center between January 2010 and December 2018 (n = 2352). Of these patients, 6.8% (161 of 2352) had an ipsilateral hip or knee implant in situ at the time of receiving surgical treatment for a PJI of the hip or knee. The following criteria led to the exclusion of 39% (63 of 161) of these patients: 4.3% (seven of 161) for incomplete documentation, 30% (48 of 161) for unavailability of full-leg radiographs, and 5% (eight of 161) for synchronous infection. With regard to the latter, per internal protocol, all artificial joints were aspirated before septic surgery, allowing us to differentiate between synchronous and metachronous infection. The remaining 98 patients were included in the final analysis. Twenty patients experienced ipsilateral metachronous PJI during the study period (Group 1) and 78 patients did not experience a same-side PJI (Group 2). We analyzed the microbiological characteristics of bacteria during the first PJI and ipsilateral metachronous PJI. Calibrated, full-length plain radiographs were evaluated. Receiver operating characteristic curves were analyzed to determine the optimal cutoff for the stem-to-stem and empty native bone distance. The mean time between the initial PJI and ipsilateral metachronous PJI was 8 ± 14 months. Patients were followed for a minimum of 24 months for any complications. Results: The risk of ipsilateral metachronous PJI in the other joint secondary to a joint implant in which PJI develops can increase up to 20% in the first 2 years after the procedure. There was no difference between the two groups in age, sex, initial joint replacement (knee or hip), and BMI. However, patients in the ipsilateral metachronous PJI group were shorter and had a lower weight (1.6 ± 0.1 m and 76 ± 16 kg). An analysis of the microbiological characteristics of bacteria at the time of the initial PJI showed no differences in the proportions of difficult-to-treat, high virulence, and polymicrobial infections between the two groups (20% [20 of 98] versus 80% [78 of 98]). Our findings showed that the ipsilateral metachronous PJI group had a shorter stem-to-stem distance, shorter empty native bone distance, and a higher risk of cement restrictor failure (p < 0.01) than the 78 patients who did not experience ipsilateral metachronous PJI during the study period. An analysis of the receiver operating characteristic curve showed a cutoff of 7 cm for the empty native bone distance (p < 0.01), with a sensitivity of 72% and a specificity of 75%. Conclusion: The risk of ipsilateral metachronous PJI in patients with multiple joint arthroplasties is associated with shorter stature and stem-to-stem distance. Appropriate position of the cement restrictor and native bone distance are important in reducing the risk of ipsilateral metachronous PJI in these patients. Future studies might evaluate the risk of ipsilateral metachronous PJI owing to bone adjacency. Level of evidence: Level III, therapeutic study.
... Enterococci are considered as difficult to treat due to limited antibiotic activity against their biofilm formation and their potential for antibiotic resistance [6,[9][10][11][12]. In general, 2-stage exchange with a long interval between the surgeries is recommended to treat PJI with difficult-to-treat microorganisms [13][14][15]. ...
Article
Full-text available
We present a case with bilateral hematogenous hip periprosthetic joint infection with Enterococci which could be treated successfully with implant retention despite chronification and partial loosening. A debridement and replacement of the modular components was carried out with replacement of a loose acetabular cup on the right side. Considering poor local infection control, antibiotic treatment was enhanced by local application of vancomycin. In the present case, treatment of chronic enterococcal periprosthetic joint infection while preserving the implants was successful despite unfavorable odds. Considering the duration of infection, causative microorganism, and loosening of one of the implants, staged exchange of both hip replacements would have been the standard procedure. This case illustrates that some concepts have to be challenged from time to time.
... As such, 12 GN PJIs were monomicrobial (40%). Pathogens, for which no biofilm-active antibiotics were available were considered difficult-to-treat (DDT) pathogens [15]. In this series, those included rifampin-resistant Staphylococci (1), fluoroquinolone-resistant GN bacteria (6), and Enterococci (2). ...
Article
Full-text available
While gram negative (GN) periprosthetic joint infections (PJI) have previously been described as difficult to treat pathogens with high rates of reinfection, limited investigations have addressed midterm outcomes and risk of infection persistence by the same pathogen. This study analyzed (1) baseline demographics, treatment strategy, and midterm outcomes of GN PJIs, as well as (2) differences in reinfection and relapse rates compared to gram positive (GP) PJIs. We identified 29 patients that were revised for 30 GN PJIs of total hip arthroplasties (THAs) between 2010 and 2020 using a university-based hip registry. Mean age was 77 years, 63% were females (19), and mean BMI was 27 kg/m2. Major causative pathogens included Escherichia coli (12), Klebsiella pneumoniae (5), Pseudomonas aeruginosa (5), and Enterobacter cloacae complex (5). Mean follow-up was 3.5 years. Study outcomes included (1) Kaplan–Meier survivorship analyses of all 30 GN PJIs, and (2) comparison of 18 two-stage exchanges for GN PJIs and 104 two-stage exchanges for GP PJIs, performed during the time from 2013 to 2017. (1) The 5-year survivorship free of recurrent PJI was 69%, and there were 7 recurrent PJIs at a mean of 2 years. There were 2 further suprafascial wound infections, resulting in a 61% survivorship free of any infection at 5-years. At a mean of 2 years, there were 7 patients with reinfection by the same GN pathogen (6 PJIs, one wound infection) as at index revision (23%). (2) Following two-stage exchange, the 5-year survivorship free of recurrent PJI (GN: 74%; GP: 91%; p = 0.072), any infection (GN: 61%; GP: 91%; p = 0.001), and reinfection by the same pathogen was significantly lower among GN PJIs (GN: 73%; GP: 98%; p < 0.001). Patients revised for GN PJIs are at increased risk of reinfection as opposed to GP infections. Affected patients must be counseled on the exceptionally high risk of infection persistence with one in four developing relapses. Therapeutic Level III.
... Quinolone-resistant Gram-negative bacteria, rifampicin-resistant Staphylococcus, Enterococcus and Candida were classified as "difficult to treat" (DTT) [37]. Methicillinresistant Staphylococci were abbreviated as "MRS", and all other organisms (including culture-negative infections and polymicrobial infection without the above-mentioned MRS or DTT) were classified as "easy to treat" (ETT) [36][37][38]. ...
Article
Full-text available
The accuracy of preoperative synovial fluid microbe detection in periprosthetic joint infection (PJI) is widely reported. However, the reliability of this diagnostic modality amongst the different joints is not yet described. We aimed to compare the concordance rate between preoperative synovial fluid and intraoperative tissue cultures in shoulder, knee and hip PJIs. A total of 150 patients who met the 2018 International Consensus Meeting criteria for shoulder, hip and knee PJI were retrospectively reviewed. This cohort was divided into three groups based on the involved joint (should, hip or knee), with 50 patients in each group. Cultures were collected and held for culture for 14 days. The overall concordance rate was 56.7%. Concordance rates between preoperative and intraoperative cultures were 60%, 56% and 54% for the knee, shoulder and hip joints, respectively. The analysis of high- or low-virulence and difficult- or not-difficult-to-treat germs did not reveal any significant differences between preoperative and intraoperative cultures in any of the groups. However, even considering the higher concordance in knee PJI, the overall discordance between preoperative and intraoperative cultures should prompt surgeons not to rely solely on preoperative synovial fluid culture data in determining appropriate treatment and antibiotics.
... There are many studies 175-180 on the grafting of NPs on a SAM, especially Ag and copper sulphide NPs, that have shown the applicability of these films thanks to their antibacterial and anti-biofilm activity against E. coli, Micrococcus luteus, S. aureus and S. epidermidis. This finding is very important, considering that these bacterial strains are often responsible for infections in breast implants [181][182][183] , central venous catheters 184 , cochlear implants 185 , endotracheal tube 186,187 , feeding tube 188 , orthopedic implants 189,190 , stents 191 and urinary catheters 192 . Multilayers can also be loaded with AgNPs. ...
Article
Full-text available
Healthcare-associated infections are a serious threat in terms of morbidity and mortality for all patients receiving healthcare. The problem is aggravated by the increasingly widespread phenomenon of antibiotic resistance, with some microorganisms now resistant to all or almost all the currently available antibiotics. Nanomaterials are compounds used by many different industrial fields and they are currently studied for their intrinsic antimicrobial properties. To date, many researchers have considered using many different nanoparticles and nanomaterials to produce surfaces and medical devices with intrinsic antimicrobial features. Many compounds have shown very interesting and effective antimicrobial capacities and could be used, in the future, to manufacture new hospital surfaces and medical devices. However, many studies have to be carried out to evaluate the effective potential use of these compounds. The aim of this paper is to review the main literature regarding this topic, focusing on the main types of nanoparticles and nanomaterials studied for this purpose.
... Infection studies on these cases found that these infections in 90% of cases are caused by staphylococcus of which half show negative coagulase test. Other pathogens causing septic arthritis after the surgery are propionibacterium acnes and enterobacters [1]. Vancomycin is considered one of the most important effective antibiotic and its effectiveness on septic arthritis treatment has been proved in several studies [4,11,17]. ...
Article
Full-text available
Introduction Joint infection after Anterior Cruciate Ligament (ACL) reconstruction is an uncommon infection which can affect joint movement and function. In this study, the impact of using antibiotic during graft preparation on the results of ACL reconstruction was investigated to examine the negative effects of antibiotic solution on graft and clinical symptoms after the surgery. Methods In this randomized clinical trial study, 80 patients were enrolled. In one group, the graft was placed in vancomycin solution (500 mg of vancomycin powder in 100 ml of normal saline) for 10–15 min during the surgery. In other group, the surgery was performed routinely and the graft was not placed in antibiotic solution. Intravenous antibiotic was given to both groups and they underwent ACL reconstruction surgery through arthroscopic transportal technique using their hamstring tendon. Symptoms and examinations of patients were evaluated for one year after the surgery. Results There was no difference between two groups in terms of knee dislocation, knee lock, pain, fever, positivity of Lachman test, Anterior drawer test and pivot-shift test, knee swelling, and movement restriction in flection and extension (P > 0.05). No infection was seen in patients. Conclusions Placing grafts in vancomycin solution does not have negative effects on graft quality and results of ACL surgery.
Article
Periprosthetic joint infection is the main social and economic problem of modern orthopedics with a recurrence rate of chronic forms of up to 23.2–31.5%. The aim of this review is to inform various specialties doctors about the features of pathogenesis, etiology and treatment of periprosthetic joint infection, which significantly differ it from of other surgical site infections. The severity of infectious complications is due to the suppression of the patient’s immune system and microbial biofilms. Surgical treatment of periprosthetic joint infection involves debridement with preservation or removal of the implants, resection arthroplasty and arthrodesis. Today, in the treatment of more than 80% of cases of chronic infection, a two-stage approach is used, which allows to restore joint function after an average of 1.0–1.5 years. An integral part of the treatment of patients is high-dose, long-term and combined antibiotic therapy, which allows you to effectively deal with the leading pathogens of periprosthetic infection Staphylococcus aureus and S. epidermidis, the share of which is 46.5–57.5%. In the conditions of growing resistance of bacteria to antibiotics, the interest of researchers in the possibilities of using bacteriophages in the complex therapy of infections of bones and joints has increased. The cost of treatment exceeds the cost of “aseptic” joint replacement by 2–24 times and is characterized by a high level of disability and mortality of patients. Taking into account the numerous factors affecting the course and effectiveness of complex treatment of patients with periprosthetic infection, a multidisciplinary approach is currently considered the main component of success. The medical and social significance and high financial costs of treating patients with periprosthetic infection indicate the need for further research and the active implementation of effective scientific developments in the practical healthcare system.
Article
Our knowledge about the fundamental aspects of biofilm biology, including the mechanisms behind the reduced antimicrobial susceptibility of biofilms, has increased drastically over the last decades. However, this knowledge has so far not been translated into major changes in clinical practice. While the biofilm concept is increasingly on the radar of clinical microbiologists, physicians, and healthcare professionals in general, the standardized tools to study biofilms in the clinical microbiology laboratory are still lacking; one area in which this is particularly obvious is that of antimicrobial susceptibility testing (AST). It is generally accepted that the biofilm lifestyle has a tremendous impact on antibiotic susceptibility, yet AST is typically still carried out with planktonic cells. On top of that, the microenvironment at the site of infection is an important driver for microbial physiology and hence susceptibility; but this is poorly reflected in current AST methods. The goal of this review is to provide an overview of the state of the art concerning biofilm AST and highlight the knowledge gaps in this area. Subsequently, potential ways to improve biofilm-based AST will be discussed. Finally, bottlenecks currently preventing the use of biofilm AST in clinical practice, as well as the steps needed to get past these bottlenecks, will be discussed.
Article
Full-text available
Aims: To investigate the outcomes of treatment of streptococcal periprosthetic joint infection (PJI) involving total knee and hip arthroplasties. Patients and methods: Streptococcal PJI episodes which occurred between January 2009 and December 2015 were identified from clinical databases. Presentation and clinical outcomes for 30 streptococcal PJIs in 30 patients (12 hip and 18 knee arthroplasties) following treatment were evaluated from the medical notes and at review. The Kaplan-Meier survival method was used to estimate the probability of infection-free survival. The influence of the biofilm active antibiotic rifampin was also assessed. Results: The infection was thought to have been acquired haematogenously in 16 patients and peri-operatively in 14. The median follow-up time for successfully treated cases was 39.2 months (12 to 75), whereas failure of the treatment occurred within the first year following treatment on every occasion. The infection-free survival at three years with 12 patients at risk was 59% (95% confidence interval 39% to 75%). Failure of the treatment was observed in ten of 22 PJIs (45%) treated with a two-stage revision arthroplasty, two of six (33%) treated by debridement and prosthesis retention, and in neither of the two PJIs treated with one-stage revision arthroplasty. Streptococcal PJI treated with or without rifampin included in the antibiotic regime showed no difference in treatment outcome (p = 0.175). Conclusion: The success of treatment of streptococcal PJI in our patient cohort was poor (18 of 30 cases, 59%). New therapeutic approaches for treating streptococcal PJI are needed. Cite this article: Bone Joint J 2017;99-B:653-9.
Article
Full-text available
Background Periprosthetic tissue cultures, sonication and synovial fluid cultures remain the gold standard for prosthetic joint infection (PJI) diagnosis. However, some 15–20% culture-negative PJI are still reported. Therefore, there is the need for other diagnostic criteria. One point of concern relative to the different definitions of PJI is as to the inclusion of the c-reactive protein (CRP) and the erythrocyte sedimentation rate (ESR) as diagnostic criteria for PJI despite them being non-specific inflammatory blood tests. PurposeThe purpose of the present study was to determine the relevance of CRP and the ESR in the diagnosis of PJI. Methods All PJI with positive cultures over a two-year period in two hospitals were reviewed. The main variables of the present study were the type of prosthesis and the CRP level. More information was recorded in those patients with normal CRP: radiographs, physical examination records and the ESR. ResultsSeventy-three patients were included in study. Pre-operative CRP levels were normal (lower than 0.8 mg/dl) in 23 patients, representing 32% of all PJI with positive cultures. Low virulence micro-organisms, 12 coagulase-negative staphylococci and four P. acnes, grew in most of them. They represented 70% of all PJI with normal CRP levels. In addition, 17 patients (23% of all PJI with positive cultures) had a normal ESR, a normal physical examination (they only presented with pain) and no clear loosening was observed in the radiographs. Conclusions Per the American Association of Orthopaedic Surgeons (AAOS) guidelines or the Musculoskeletal Infection Society (MSIS), 23% of the patients in the present study with PJI would never have been identified. Blood inflammatory markers such as the CRP level and ESR may not be accurate as diagnostic tools in PJI, particularly to identify low-grade and chronic PJI.
Article
Full-text available
Introduction: Besides CRP in serum, white cell counts and cultures of synovial fluid are routinely used to detect periprosthetic joint infections. But the sensitivities of these parameters do vary from 12 to 100 %. In two stage revision arthroplasty before the second stage surgeons have to decide if reimplantation is justified. Therefore, we investigated the value of cultures and white cell count from the synovial fluid with a polymethyl methacrylate spacer in place and CRP in serum before reimplantation to detect persistent infection in a standardized setting. Methods: 115 patients with a two-stage revision hip or knee arthroplasty were included in this study. All patients had an antibiotic loaded polymethylmethacrylate spacer. Retrospectively synovial cultures, white blood count in synovial fluid and CRP in serum were assessed before reimplantation. Results: The sensitivity of the synovial cultures was 5 % (95 % CI 0.13-24.87), with a specificity of 99 % (95 % CI 94.27-99.97). For white blood count in synovial fluid the sensitivity was 31.3 %, specificity was 39.1 %. Sensitivity for CRP in serum was 42.10 %, specificity was 84.21 %. Conclusion: Cultures from synovial fluid and white blood count in synovial fluid and CRP seem to be uncertain parameters to exclude persistent infection. We do not recommend joint aspiration before reimplantation anymore. Further research is necessary to find other markers to confirm or exclude persistent infection.
Article
AIMS: The aim of this study was to identify the incidence of positive cultures during the second stage of a two-stage revision arthroplasty and to analyse the association between positive cultures and an infection-free outcome. PATIENTS AND METHODS: This single-centre retrospective review of prospectively collected data included patients with a periprosthetic joint infection (PJI) of either the hip or the knee between 2013 and 2015, who were treated using a standardised diagnostic and therapeutic algorithm with two-stage exchange. Failure of treatment was assessed according to a definition determined by a Delphi-based consensus. Logistic regression analysis was performed to assess the predictors of positive culture and risk factors for failure. The mean follow-up was 33 months (24 to 48). RESULTS: A total of 163 two-stage revision arthroplasties involving 84 total hip arthroplasties (THAs) and 79 total knee arthroplasties (TKAs) were reviewed. In 27 patients (16.6%), ≥ 1 positive culture was identified at re-implantation and eight (29.6%) of these subsequently failed compared with 20 (14.7%) patients who were culture-negative. The same initially infecting organism was isolated at re-implantation in nine of 27 patients (33.3%). The organism causing re-infection in none of the patients was the same as that isolated at re-implantation. The risk of the failure of treatment was significantly higher in patients with a positive culture (odds ratio (OR) 1.7; 95% confidence interval (CI) 1.0 to 3.0; p = 0.049) and in patients with a higher Charlson Comorbidity Index (OR 1.5; 95% CI 1.6 to 1.8; p = 0.001). CONCLUSION: Positive culture at re-implantation was independently associated with subsequent failure. Surgeons need to be aware of this association and should consider the medical optimisation of patients with severe comorbidities both before and during treatment.
Article
Despite the lack of validation, synovial aspiration remains a common practice during 2-stage septic revision total knee arthroplasty (TKA). The goal of this study was to investigate the diagnostic validity of synovial polymethylmethacrylate (PMMA) spacer aspiration of temporary knee arthrodesis to detect persistent periprosthetic joint infection before TKA reimplantation. This retrospective cohort study included 73 consecutive patients who underwent 2-stage septic revision TKA according to a standard protocol. After explantation surgery, including temporary arthrodesis with an intramedullary stabilized PMMA spacer, all patients had synovial aspiration 2 weeks before reimplantation to exclude persistent periprosthetic joint infection. Patients had a 2-week antibiotic holiday before aspiration. Sensitivity and specificity of the synovial PMMA spacer joint aspiration for the detection of periprosthetic joint infection were determined and referenced against intraoperative microbiologic and histologic samples obtained at second-stage surgery. Sensitivity of the synovial PMMA spacer aspiration was 21%. Because of poor diagnostic validity, synovial PMMA spacer aspiration cannot be recommended for routine exclusion of persistent periprosthetic joint infection before TKA reimplantation. Therefore, exclusion of persistent periprosthetic joint infection should be supplemented by other diagnostic methods, and it is not necessary to delay TKA reimplantation for PMMA spacer aspiration. [Orthopedics. 201x; xx(x):xx-xx.].
Article
Many infections of the musculoskeletal system are biofilm infections that develop on non-living surfaces. Microorganisms adhere either on dead bone (sequesters) or implants. As a rule for a curative concept, chronic osteomyelitis or implant-associated bone infection must be treated with a combination of surgery and antimicrobial therapy. If an implant is kept in place, or a new device is implanted before complete healing of infection, a biofilm-active antibiotic should be used. Rifamycins are active against biofilms of staphylococci, and fluoroquinolones against those of Gram-negative bacilli. In this review, the management of chronic osteomyelitis, periprosthetic joint infection and implant-associated osteomyelitis of long bones is presented.
Article
Research that leads to better strategies to diagnose and treat prosthetic joint infection (PJI) is critical because PJI is a devastating complication of total knee arthroplasty. A key to the diagnosis and management of PJI is defining the microbiology of PJI and improving the medical management of PJI utilizing both systemic and local antimicrobial therapy. In this review, the author will present his opinions on future research needs as they relate to the microbiology of PJI, including antimicrobial resistance and the antimicrobial treatment of PJI. This paper summarizes a presentation given at a recent multidisciplinary research conference entitled "Strategies to improve total knee arthroplasty" sponsored by the Knee Society. It was a part of a session entitled "Periprosthetic Joint Infection."
Article
Infections involving orthopaedic surgical implants present unique challenges when compared with infections that do not involve implants. Microorganisms have a high affinity for adhering to foreign materials commonly used in orthopaedics, including cobalt-chromium, titanium, polyethylene, and polymethylmethacrylate (PMMA) cement. When bacteria adhere to these surfaces, they can form a complex structure surrounded by a self-generated extracellular polymeric substance (EPS) matrix formed by multiplex agents of biopolymers consisting of proteins, polysaccharides, lipids, nucleic acids, and humic substances1-3. The term “biofilm” is commonly used to describe this network of microorganisms, a term popularized by Dr. J. William Costerton et al. in 19784. Biofilms are formed by a confluence of bacteria commonly encountered in orthopaedic infections. Up to 65% of bacterial infections are caused by biofilm-producing organisms5. Staphylococci, specifically Staphylococcus aureus ( S. aureus ) and Staphylococcus epidermidis ( S. epidermidis ), are the most common biofilm-forming bacteria found in orthopaedics, and, when combined with Pseudomonas aeruginosa ( P. aeruginosa ), they represent nearly 75% of biofilm infections observed in medical devices6. Propionibacterium acnes ( P. acnes ), an organism commonly found in shoulder infections, has also been shown to form biofilm. Biofilms can be composed of a single organism or can be polymicrobial; polymicrobial biofilms are more difficult to eradicate7. Once bacteria adhere to the surface of implants, they may replicate and may form a complex network of microorganisms that communicate with one another via cell-to-cell signaling that facilitates the participation of bacteria in quorum sensing7. Quorum sensing serves as an elementary endocrine system whereby bacteria sense the local cell population density and regulate gene expression by releasing extracellular molecules to facilitate synchronized changes in the bacteria within the biofilm. These transcriptional changes can occur with the exchange of plasmids between bacteria, which can confer genes …
Article
The objective of this study was to develop a prospectively applicable method for classifying comorbid conditions which might alter the risk of mortality for use in longitudinal studies. A weighted index that takes into account the number and the seriousness of comorbid disease was developed in a cohort of 559 medical patients. The 1-yr mortality rates for the different scores were: "0", 12% (181); "1-2", 26% (225); "3-4", 52% (71); and "greater than or equal to 5", 85% (82). The index was tested for its ability to predict risk of death from comorbid disease in the second cohort of 685 patients during a 10-yr follow-up. The percent of patients who died of comorbid disease for the different scores were: "0", 8% (588); "1", 25% (54); "2", 48% (25); "greater than or equal to 3", 59% (18). With each increased level of the comorbidity index, there were stepwise increases in the cumulative mortality attributable to comorbid disease (log rank chi 2 = 165; p less than 0.0001). In this longer follow-up, age was also a predictor of mortality (p less than 0.001). The new index performed similarly to a previous system devised by Kaplan and Feinstein. The method of classifying comorbidity provides a simple, readily applicable and valid method of estimating risk of death from comorbid disease for use in longitudinal studies. Further work in larger populations is still required to refine the approach because the number of patients with any given condition in this study was relatively small.