Next Article in Journal
Estimates of Dietary Exposure to Antibiotics among a Community Population in East China
Next Article in Special Issue
Microbiological Trends and Antibiotic Susceptibility Patterns in Patients with Periprosthetic Joint Infection of the Hip or Knee over 6 Years
Previous Article in Journal
ICU-Associated Gram-Negative Bloodstream Infection: Risk Factors Affecting the Outcome Following the Emergence of Colistin-Resistant Isolates in a Regional Greek Hospital
Previous Article in Special Issue
Impact of High-Dose Anti-Infective Agents on the Osteogenic Response of Mesenchymal Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Orthopaedic Implant-Associated Staphylococcal Infections: A Critical Reappraisal of Unmet Clinical Needs Associated with the Implementation of the Best Antibiotic Choice

1
Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
2
SSD Clinical Pharmacology, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
3
Bacteriology Unit, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy
4
Department of Anaesthesia and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy
5
UOSD Microbiologia Arezzo PO San Donato, Azienda Usl Toscana Sud Est, 52100 Arezzo, Italy
6
Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
7
Infectious and Tropical Diseases Unit, Careggi University Hospital, 50134 Florence, Italy
8
Neurointensive Care Unit, Department of Anesthesiology, Careggi University Hospital, 50134 Florence, Italy
9
Agenzia Regionale di Sanità della Toscana, 50141 Florence, Italy
10
SOD Microbiologia e Virologia, Careggi University Hospital, 50134 Florence, Italy
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(3), 406; https://doi.org/10.3390/antibiotics11030406
Submission received: 14 February 2022 / Revised: 14 March 2022 / Accepted: 16 March 2022 / Published: 17 March 2022

Abstract

:
Infections associated with orthopaedic implants represent a major health concern characterized by a remarkable incidence of morbidity and mortality. The wide variety of clinical scenarios encountered in the heterogeneous world of infections associated with orthopaedic implants makes the implementation of an optimal and standardized antimicrobial treatment challenging. Antibiotic bone penetration, anti-biofilm activity, long-term safety, and drug choice/dosage regimens favouring outpatient management (i.e., long-acting or oral agents) play a major role in regards to the chronic evolution of these infections. The aim of this multidisciplinary opinion article is to summarize evidence supporting the use of the different anti-staphylococcal agents in terms of microbiological and pharmacological optimization according to bone penetration, anti-biofilm activity, long-term safety, and feasibility for outpatient regimens, and to provide a useful guide for clinicians in the management of patients affected by staphylococcal infections associated with orthopaedic implants Novel long-acting lipoglycopeptides, and particularly dalbavancin, alone or in combination with rifampicin, could represent the best antibiotic choice according to real-world evidence and pharmacokinetic/pharmacodynamic properties. The implementation of a multidisciplinary taskforce and close cooperation between microbiologists and clinicians is crucial for providing the best care in this scenario.

1. Introduction

Infections associated with orthopaedic implants represent a major health concern characterized by a remarkable incidence of morbidity and mortality [1,2]. The growth of elderly and immunocompromised populations, coupled with the great efficacy of joint replacement interventions in improving patients’ quality of life, have recently led to an increased incidence of infections associated with orthopaedic implants [3,4]. While in patients undergoing primary joint replacement, the infection rate during the first two years is approximatively less than 1–2%, the infections rate after revision surgery may rise up to 40%. Similarly, a wide difference in the infection rate of internal fixation devices exists, considering that higher infective risk occurs after the fixation of open fractures (up to 30%) compared to closed fractures (less than 2%) [1].
Infections associated with orthopaedic implants, especially prosthetic joint infections (PJIs), are commonly classified as early-onset, delay-onset, and late-onset infections, with infections occurring in the first 3 months, between 3 and 12–24 months, or after 24 months, respectively [1,4]. Causative pathogens consist of virulent microorganisms (e.g., Staphylococcus aureus, Enterococcus spp., Gram-negative bacilli) in case of early-onset infections, while low-virulence pathogens, e.g., coagulase-negative staphylococci (CoNS), Propionibacterium acnes, are mostly involved in delayed- and late-onset infections [1,4]. Furthermore, it is important to recognize the resistance pattern of involved pathogens, considering that methicillin-resistant S. aureus (MRSA) strains may account for 25–50% of isolates [5]. Notably, S. aureus represent the leading causative pathogen in infections associated with orthopaedic implants, exhibiting remarkable virulence, particularly in patients with multiple comorbidities, and being involved in both early-onset and delayed- or late-onset cases [2]. Concomitant bacteraemia is retrieved in up to 60% of patients with infections associated with orthopaedic implants, with higher rates in patients treated with debridement with prosthesis retention than in patients with resection arthroplasty [2].
The wide variety of clinical scenarios encountered in the heterogeneous world of infections associated with orthopaedic implants makes the implementation of an optimal and standardized antimicrobial treatment in terms of drug choice, dosage regimen, and treatment duration challenging. While pathophysiological conditions exhibit a lower relevance compared to critically ill patients in the definition of the antimicrobial therapy puzzle [6], antibiotic bone penetration [7,8], anti-biofilm activity [9], long-term safety, and drug choice/dosage regimen favouring outpatient management (i.e., long-acting or oral agents), especially in the current COVID-19 era [10], play a major role in the chronic evolution of these infections (Figure 1).
Furthermore, the coordinated approach of a multidisciplinary task force including medical and surgical specialists (namely infectious disease consultants, clinical microbiologists, MD clinical pharmacologists, orthopaedic surgeons, intensive care physicians) could maximize treatment efficacy and improve clinical outcomes, as reported in other settings [11,12].
The Tuscany region has implemented an innovative organization called Rete AID (where A, I, and D refer to Antimicrobial Stewardship, Infection Control, and Diagnostic Stewardship, respectively) [13] for the systemic approach to fight microbiological threats. This organization can access a large information set provided by the collaborative network of clinical microbiology laboratories in the region, using the Regional Health Agency ARS (SMART network, Microbiological Surveillance and Antibiotic Resistance in Tuscany). In this context, a multidisciplinary task force, called Network Lab, including microbiologists, infectious disease specialists, orthopaedic surgeons, cardiac surgeons, and intensive care physicians, was implemented in order to develop a best practice model in the diagnosis and management of infections associated with implantable devices. In Tuscany, the rate of early-onset and delay-/late-onset infections associated with orthopaedic implants accounted for less than 1% and approximatively 2% of infections, respectively, while the rate of MRSA isolates decreased in recent years to below 30% [14].
Although several guidelines concerning the management of bone and joint infections have been issued in the last decade [15,16,17], a comprehensive overview focusing on the rational choice of available therapeutic alternatives from a microbiological and pharmacological standpoint is lacking [15].
This multidisciplinary opinion article aims to summarize evidence supporting the use of the different anti-staphylococcal agents in terms of microbiological and pharmacological optimization and to provide a useful guidance for clinicians in the management of patients affected by infections associated with orthopaedic implants.

2. Results

2.1. Issues in Microbiological Diagnosis of Infections Associated with Orthopaedic Implants

The microbiological diagnosis of orthopaedic implant-associated infections represents a crucial aspect of the management of these challenging conditions. As stated in the international guidelines [15], the detection of two or more positive cultures from intraoperative specimens, or a combination of preoperative aspiration and intraoperative cultures that yield the same organism (indistinguishable based on common laboratory tests including genus and species identification or common antibiogram) represents a definitive evidence of infection, providing acceptable sensitivity and specificity without requiring an impractical amount of tissue specimens to be processed by the laboratory [18]. Additionally, the growth of a virulent microorganism (e.g., S. aureus) in a single specimen of a tissue biopsy or synovial fluid may also be sufficient for the diagnosis of implant-associated infection [15]. Conversely, one of multiple tissue cultures or a single aspiration culture that yields an organism that is a common contaminant (e.g., CoNS, Propionibacterium acnes) should not necessarily be considered evidence of definite implant-associated infection and should be evaluated in the context of other available evidence [15].
Notably, microbiological diagnosis is also crucial for the identification of causing pathogens and their susceptibility, allowing the implementation of a targeted antibiotic therapy. At least 3, and optimally 5 or 6, periprosthetic intraoperative samples from the most suspicious areas of tissue as deemed by the orthopaedic surgeon should be obtained for aerobic and anaerobic culture for the optimal diagnosis of implant-associated infection, considering that the collection of fewer than 5–6 specimens leads to a decrease in the sensitivity of the culture as a diagnostic test [18,19]. The optimal duration of incubation of periprosthetic tissue specimens is currently unknown, but prolonged incubation of up to 14 days may help the isolation and identification of slow-growing low-virulent pathogens (e.g., Propionibacterium spp.) [20]. The implementation of novel processing techniques may also improve pathogen identification [21]. Sonication of the explanted prosthesis, with the aim to dislodge pathogens from the surface, followed by subsequent aerobic and anaerobic culture may improve the diagnostic sensitivity compared to traditional tissue cultures, increasing from 60.8% to 78.5% [19,20,21,22,23].
Novel molecular testing, e.g., multiplex polymerase chain reaction (PCR) performed on joint fluid aspirate may provide a rapid and accurate diagnosis of implant-associated infections, considering that higher diagnostic accuracy for the detection of PJIs and higher detection of low-virulence pathogens (e.g., CoNS, Propionibacterium spp.) compared to traditional cultures were reported [2,24,25].

2.2. Role of Bacterial Biofilm

The presence of biofilm plays a crucial role in the pathogenesis of orthopaedic implant-associated infections [2,9]. Both virulent bacteria (e.g., S. aureus) and opportunistic pathogens (e.g., CoNS) are able to form biofilm [9]. The formation and maturation of bacterial biofilm protects the pathogen from both the host immune response and antibiotics, making the treatment and eradication of this condition extremely challenging, considering that biofilm-embedded bacteria are 100–1000 times less susceptible to antibiotics [9]. Furthermore, biofilm-embedded bacteria are usually in a slow-growing or stationary state, leading to the development of bacterial subpopulations persisting in the biofilm in a dormant metabolic state that is highly resistant to antibiotics, known as small colony variants (SCVs) [26,27]. SCVs are characterized by unusual morphological appearance and biochemical reactions, often remaining undetected or misdiagnosed [27]. Additionally, they may exhibit an unstable phenotype, possibly reverting to the highly virulent and rapidly growing form [26,27].
An ideal anti-biofilm antibiotic should provide bactericidal activity, efficacy against bacteria in a stationary phase and biofilm-producing pathogens, and the ability to penetrate within biofilm as necessary requirements (Figure 2) [9].
In order to assess the anti-biofilm activity of a certain agent, classic antibiotic susceptibility tests providing the MIC required to define clinical breakpoints and PK/PD targets are inadequate [28]. Consequently, other pharmacodynamic parameters were developed to quantify antimicrobial activity against biofilm-producing pathogens, namely the minimal biofilm inhibitory concentration (MBIC), the minimal biofilm bactericidal concentration (MBBC), and the minimal biofilm eradication concentration (MBEC) [28]. However, the MBECs for common anti-staphylococcal agents are between 10- and 8000-fold higher than the MICs, thus making the choice of the best antibiotic strategy extremely challenging [9].

2.3. Real-World Use of Traditional and Novel Anti-Staphylococcal Agents for the Management of Infections Associated with Orthopaedic Implants

2.3.1. Novel Lipoglycopeptide (Dalbavancin, Oritavancin, and Telavancin)

Several real-world experiences [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57] document the efficacy of novel lipoglycopeptide, namely dalbavancin, oritavancin, and telavancin, in the management of infections associated with orthopaedic implants (Table S1).
Currently, the evidence supports the use of dalbavancin in this scenario, with an overall clinical success rate above 75–80% [10,29]. Specifically, nine prospective and retrospective studies [30,31,32,33,34,35,36,37,38], one case series [39], and four case reports [40,41,42,43] assessed the efficacy and safety of dalbavancin for the treatment of PJIs and implant-associated infections. Morata et al. [30] reported the largest sample size, retrospectively studying 64 patients with bone and joint infections, of which 45 and 19 were affected by implant-associated infections and osteomyelitis/PJIs, respectively. Staphylococcus epidermidis was isolated in almost half of the cases. A clinical success or improvement was respectively reported in 97.7% and 89.5% of subjects with implant-associated infections and osteomyelitis/PJIs. Notably, a wide heterogeneity in the dalbavancin dosing schedule and treatment duration was noted. Overall, the rate of adverse events (AEs) ranged from 2% to 13%, most of which were classified as non-serious.
Real-world evidence documenting the efficacy of oritavancin is currently limited to a retrospective study [44] and a case series [45] including only four patients affected by infections associated with orthopaedic implants. Overall, a clinical cure and/or improvement was documented in 75% of cases, while no AEs were reported.
A retrospective study [46] and a case series [47] assessed the efficacy and safety of telavancin for the management of infections associated with orthopaedic implants. Notably, Sims et al. [46] retrospectively studies 57 patients, of which 30 were affected by osteomyelitis with prosthetic materials and 27 were affected by PJIs. S. aureus accounted for more than half of cases. Overall, the clinical cure was 75% and 84.6% in the osteomyelitis and PJIs group, respectively. AEs were reported in approximatively 20% of cases, of which 2.1% was classified as serious.
In several in vitro experiences, all of the three novel lipoglycopeptides showed high anti-biofilm activity against Staphylococci and Enterococci clinical isolates. Both lower minimum biofilm bactericidal concentrations (MBBCs) and minimal biofilm eradication concentrations (MBECs) were found when compared to treatment with vancomycin, linezolid, tedizolid, and cloxacillin [48,57]. Additionally, an additive or synergistic anti-biofilm activity was found for dalbavancin and oritavancin in combination with rifampicin [53,56].
Notably, both dalbavancin and oritavancin exhibited peculiar pharmacokinetic properties, consisting in a long half-life (higher than 7–14 days) and high bone penetration [58], thus allowing for favourable dosing regimens in terms of the outpatient management of infections associated with orthopedic implants. Specifically, preclinical studies showed that for both dalbavancin and oritavancin, bone concentrations were well above MIC90 of susceptible pathogens for up to 7–14 days [59,60]. Conversely, telavancin exhibited a short half-life (7–9 h); thus, daily administration is required [58]. Furthermore, studies assessing telavancin bone penetration is lacking.

2.3.2. Novel Anti-Staphylococcal Cephalosporins (Ceftaroline and Ceftobiprole)

Real-world evidence concerning the use of novel anti-MRSA cephalosporins, namely ceftaroline and ceftobiprole, in the management of infections associated with orthopaedic implants is currently limited (Table S2).
A retrospective cohort study including only 19 patients, of which 16 were affected by implant-associated infections, assessed the efficacy of ceftaroline in this scenario [61]. Overall, a clinical cure was achieved in 68.4% of cases at end of treatment, but was maintained at one year in only approximatively 10% of patients. Furthermore, a remarkable rate of AEs was reported (more than 20%). Ceftaroline exhibited promising in vitro activity against S. aureus and Staphylococcus epidermidis strains retrieved in PJIs, with MIC90/50 below 0.5 mg/L [62]. Notably, ceftaroline exerted a bactericidal activity against both MSSA and MRSA biofilms, although a lack of complete biofilm eradication was found [63]. An additive and/or synergic effect with daptomycin and rifampicin was reported in preclinical/in vitro models of infections associated with medical devices, including PJIs [64,65].
Real-world evidence concerning ceftobiprole administration for the management of bone and joint infections is limited to a single retrospective study including nine patients [66]. The findings were promising in terms of a clinical cure (77.8%) and microbiological eradication rate (88.9%). Notably, no AEs were reported. Furthermore, preclinical/in vitro models showed an effective anti-biofilm activity and effective bone penetration compared to vancomycin or linezolid [67,68].

2.3.3. Daptomycin

Three comparative studies [69,70,71], one prospective observational study [72], five retrospective cohort studies [73,74,75,76,77], and two case reports [78,79] investigated the efficacy and safety of daptomycin in the management of infections associated with orthopaedic implants, for a total of 277 included patients (Table S3).
In a randomized controlled trial, Byren et al. [69] compared 49 patients affected by PJIs managed with two-stage revision receiving daptomycin 6–8 mg/kg/day with 26 patients receiving vancomycin, teicoplanin, or semisynthetic penicillins. Higher clinical cure (59.6% vs. 38.1%) and microbiological eradication (51.1% vs. 38.1%) rates were found in patients treated with daptomycin compared to other regimens. No difference in terms of AE rate leading to treatment discontinuation was reported (12.2% vs. 16.0%). In a retrospective comparative study including 341 patients affected by PJIs, Carli et al. [70] found that daptomycin use (n = 77) was not associated with better clinical outcome in patients undergoing either DAIR (OR 1.70; 95% CI 0.62–4.65) or two-stage exchange (OR 0.58; 95% CI 0.27–1.26) treatment when compared to other regimens. In a retrospective matched case-control study, Joseph et al. [71] reported no difference in either clinical cure (85% vs. 90%; p = 0.63) or relapse rate (10% vs. 10%; p = 0.99) in 20 patients receiving daptomycin for the management of PJIs compared to 20 cases treated with vancomycin. Notably, although no significant difference was reported in overall AE rate (20% vs. 30%; p = 0.47), cases receiving vancomycin experienced a higher rate of discontinuation due to severe AEs (25% vs. 0%; p = 0.02).
Overall, a remarkable clinical cure rate, ranging from 50.0% to 100.0%, was found in different retrospective studies and case reports [72,73,74,75,76,77,78,79] including patients affected by PJIs, mainly due to MRSA or MRSE, and managed with daptomycin alone or in combination with rifampicin. AEs were reported in up to 20% of patients.
Notably, a phase I study [80] including 16 arthroplasty patients found a mean daptomycin bone penetration rate of 14.1 ± 11.9% after a dose of 8 mg/kg. Bone concentrations were well above MIC90 of pathogens susceptible to daptomycin.
Both in vitro and an animal foreign-body infection models [81,82] found promising results with the association of high-dose daptomycin with rifampicin or ceftaroline in biofilm-producing infections caused by MRSA or VRE. However, Molina Manso et al. [83] reported that daptomycin was not effective against biofilm produced by S. aureus and Staphylococcus epidermidis isolates in PJIs, with minimum biofilm eradication concentrations (MBECs) significantly above the minimum inhibitory concentrations (>1024 mg/L).

2.3.4. Linezolid and Tedizolid

Four prospective observational studies [84,85,86,87], two retrospective matched case-control studies [88,89], and six retrospective cohort studies [90,91,92,93,94,95] investigated the efficacy and the safety of linezolid in the management of infections associated with orthopaedic implants, for a total of 365 included patients (Table S4).
The overall clinical cure or remission rate was 79.7%, ranging from 68.2% to 100.0%. Notably, the overall AE rate in patients receiving long-term treatment with linezolid was remarkable, and AEs leading to linezolid discontinuation was reported in up to 14.3% of patients. Furthermore, no significant difference in clinical outcome was reported in patients affected by PJIs managed with DAIR receiving linezolid in monotherapy or in combination with rifampicin [90]. Similarly, no difference in clinical remission was found in 18 patients affected by orthopaedic implant-associated infections (of which 11 were PJIs) receiving linezolid in association with rifampicin, compared to 18 matched patients treated with cotrimoxazole plus rifampicin (89.3% vs. 78.6%; p = 0.47) [88].
The largest real-world experience was reported in the prospective observational study by Soriano et al. [84], in which 85 patients affected by orthopaedic device infections (69 PJIs, mostly due to MRSA or MRSE) receiving linezolid 600 mg q12h for a median of 60 days. The overall clinical remission rate was 68.2%, with 10.5% of patients experiencing at least one AE.
Real-world evidence concerning the use of tedizolid for the management of orthopaedic device-associated infections is limited to a retrospective multicenter study [96] and a single case report [97] (Table S4). Benavent et al. [96] retrospectively studied 51 cases of bone and joint infections treated with tedizolid at a dosage of 200 mg/day for a median of 29 days. Coagulase-negative Staphylococci were isolated in approximatively half of the cases. A total of 29 orthopaedic implant-associated infections were included, 17 of which were PJIs, with a clinical cure rate of 83% (76% in patients affected by PJIs), while the overall AE rate was limited to 6% of patients (only mild AEs were reported).
Notably, in preclinical models, both linezolid and tedizolid were ineffective in eradicating mature biofilm formed by MRSA and MSSA, although a certain activity in preventing biofilm formation was noted, particularly for tedizolid [98,99].
Two different phase I studies [100,101] showed that linezolid at a dosage of 600 mg achieved effective bone and synovial concentrations that were at least double the MIC90 for both Staphylococci and Streptococci, in both patients undergoing joint replacement or affected by orthopaedic implant-associated infections.

2.3.5. Vancomycin and Teicoplanin

Several guidelines recommended vancomycin as a first-line therapy in the management of prosthetic joint infections caused by oxacillin-resistant Staphylococci or penicillin-resistant Enterococci [15,17]. Different studies assessed the efficacy and the safety of novel agents (namely daptomycin and dalbavancin) compared to vancomycin in the management of orthopaedic implant-associated infections. As previously mentioned, no significant difference in the clinical outcome was found between daptomycin and vancomycin in the management of patients undergoing both DAIR or two-stage exchange [69,70,71], although a significantly lower occurrence of therapy discontinuation due to AEs was found for daptomycin [71]. Similarly, no difference in clinical cure at 1 year was found in 80 patients affected by osteomyelitis when randomized to dalbavancin or vancomycin [102], as well as in a retrospective case-control study including 215 patients (of which 102 were osteoarticular infections) receiving dalbavancin or vancomycin [103]. However, a significantly lower rate of AEs was reported in patients managed with dalbavancin compared to vancomycin [103]. No studies comparing vancomycin and linezolid were found in this scenario.
Notably, vancomycin showed poor activity in both preventing biofilm formation and in eradicating mature biofilm of the staphylococci and enterococci strains in different in vitro models when compared to novel lipoglycopeptides, with MBIC90 and MBBC90 higher than 1024 mg/L [48,51,55,57].
Phase I studies [104,105] assessing the bone penetration of vancomycin in patients affected by osteomyelitis or undergoing prosthetic joint replacement found a limited penetration in both cancellous (bone/serum ratio of 0.21) and cortical bone (mean bone/serum ratio of 0.3 ± 0.12). Moreover, Bue et al. [105] found a weaker bone penetration pattern in patients undergoing prosthetic joint replacement, with cancellous and cortical bone concentrations well below the MIC50 and MIC90 for Staphylococci, Streptococci, and Enterococci.
Teicoplanin was recommended as a first-line therapy for the management of orthopaedic implant-associated infections caused by methicillin-resistant Staphylococci, and as an alternative agent for multi-susceptible Streptococci and Enterococci in case of hypersensitivity to penicillin [106]. Peeters et al. [107] assessed 65 patients affected by bone and joint infections caused by S. aureus (17% MRSA) treated with teicoplanin alone or in combination therapy (fluoroquinolones, 45%, and rifampicin, 25%), of which 69% were classified as device-related infections. Teicoplanin was administered at a median dosage of 5.7 mg/kg/day after a loading dose consisting of five injections 12 h apart. Overall, clinical failure was found in 41.5% of patients after a median follow-up of 91 weeks, while AEs were reported in 10% of cases. Pavoni et al. [108] retrospectively studied 34 patients affected by PJIs, of which 22 received teicoplanin (mainly in combination with fluoroquinolones or rifampicin). Clinical improvement in the absence of relapse was documented in 61.9% of cases. No AEs were reported.
Interestingly, Claessens et al. [109] found that both teicoplanin and vancomycin were not effective in eradicating S. epidermidis biofilms, although the combination therapy with rifampicin improved the killing efficacy in vitro. Notably, different phase I studies found a teicoplanin bone/serum ratio ranging from 0.15 to 0.85 [110,111,112]. Furthermore, Lazzarini et al. [113] found a bone average concentration ranging from 0.9 to 2.94 mg/L in five patients undergoing total knee arthroplasty receiving 800 mg intravenous teicoplanin 2.5 h before surgery.

2.3.6. Rifampicin

Several in vitro, animal, and clinical studies have documented the benefit of rifampicin in patients with staphylococcal orthopaedic implant-associated infections, as previously summarized in a recent review [114]. Furthermore, both in retrospective and prospective comparative studies, a significantly higher clinical cure rate was found in patients affected by staphylococcal PJIs undergoing DAIR and managed with rifampicin when compared to antibiotic regimens without rifampicin (80%-95% vs. 14%-68%) [114,115]. Notably, a randomized controlled trial [116] found a significantly greater clinical cure rate in 12 patients affected by staphylococcal PJIs treated with ciprofloxacin plus rifampicin when compared to 12 receiving ciprofloxacin alone (100% vs. 58%; p = 0.02). AEs were found in five cases receiving combination therapy with rifampicin and in one case treated with ciprofloxacin alone. Conversely, in a recent multicenter randomized controlled trial, Karlsen et al. [117] reported no significant differences in the remission rate between 23 patients randomized to combination therapy with rifampicin compared to 25 cases receiving standard antimicrobial therapy (cloxacillin or vancomycin) alone (74% vs. 72%; p = 0.88), although several issues with study design and conduction emerged [115].
As previously reported [53,55,56,81,82,99,114,115,118,119], preclinical evidence demonstrated the effective anti-biofilm activity of rifampicin in staphylococcal bone and joint infection models in association with dalbavancin, oritavancin, daptomycin, linezolid, fosfomycin, and fluoroquinolones.
In phase I studies, a rifampicin bone/serum ratio of 0.2–0.5 was found, with cortical and cancellous bone concentrations well above the MIC50/90 for S. aureus [7,120,121].

2.3.7. Fluoroquinolones

Fluoroquinolones (i.e., levofloxacin, moxifloxacin, or ciprofloxacin), in association with rifampicin, are recommended as a first-line therapy for extended suppression (3–6 months) of staphylococcal PJIs [15]. A summary of the studies investigating combination therapy between fluoroquinolones and rifampicin in this scenario is provided in Table S5.
Lora-Tamayo et al. [122] randomized 63 patients affected by staphylococcal PJIs managed with DAIR to receive a combination therapy of levofloxacin plus rifampicin for 8 weeks (short duration; 33 patients) or 3–6 months (long duration; 30 patients). After a median follow-up of 540 days, no difference in clinical cure rate was found between the two regimens in both intention-to-treat (58% vs. 73%; difference—15.7%, 95% CI −39.2% to 7.8%) and per-protocol analysis (95% vs. 91.7%; difference—3.3%, 95% CI −11.7% to 18.3%). Overall, AEs were reported in 15.7% of patients. Nguyen et al. retrospectively assessed 154 patients affected by PJIs (48.1% caused by CoNS) treated with a combination therapy of levofloxacin and rifampicin. After a mean follow-up of 55.6 months, an overall clinical cure rate of 82.5% was reported, although the occurrence of AEs was remarkable (31.2% and 8.4% attributable to rifampicin and levofloxacin, respectively) [123]. In a retrospective case-control study, Wouthuyzen-Bakker et al. [124] found no significant difference in clinical cure rate in 40 patients affected by PJIs due to MSSA managed with DAIR and treated with levofloxacin plus rifampicin when compared to 18 subjects receiving moxifloxacin plus rifampicin (89% vs. 87.5%; p = 0.89). Similarly, Fily et al. [125] reported a high clinical cure rate (78.3%) in 23 patients affected by non-staphylococcus PJIs treated with moxifloxacin plus rifampicin, although at least one AE occurred in 30.4% of cases.
Interestingly, conflicting evidence emerged from preclinical studies. While different models confirmed the high efficacy of the rifampicin-levofloxacin combination against biofilm-embedded bacteria, while also preventing the selection of resistant mutants that was observed with rifampin alone [99,119], an in vitro study including ten MSSA strains derived from PJIs showed that the addition of rifampicin to levofloxacin did not improve its performance. Additionally, an increase in small colony variants was observed in the presence of rifampicin [126].
Notably, high levofloxacin penetration was found in patients undergoing bone surgery, with a mean bone/serum ratio of approximatively 50% and 100% for cortical and cancellous bone, respectively [127,128]. Mean cortical and cancellous bone concentrations were well above the MIC90 for S. aureus and Streptococci. Similarly, a mean bone/serum ratio of approximatively 100% was found for moxifloxacin after a single administration of 400 mg before joint replacement surgery, with both cortical and cancellous bone concentrations well above the MIC90 for MSSA [120,129]. Ciprofloxacin bone penetration was not directly assessed in patients undergoing joint replacement surgery or affected by bone and joint infections. However, data collected during other surgical procedures documented the achievement of effective ciprofloxacin bone concentrations after a dosage of 400 mg iv or 750 mg oral, exceeding the clinical breakpoints of Gram-positive and -negative pathogens commonly retrieved in PJIs [7].

2.3.8. Other Anti-Staphylococcal Agents (Fosfomycin, Cotrimoxazole, Tetracyclines, Clindamycin)

Fosfomycin, cotrimoxazole, tetracyclines, and clindamycin are recommended as alternative agents for the management of orthopaedic implant-associated infections in acute or chronic long-term scenarios [15].
Fosfomycin as part of a combination antimicrobial therapy at a dosage of 4–24 g/day was found to be highly effective in the management of bone and joint infections mainly caused by S. aureus, with an overall clinical cure rate of 82.2% and a limited occurrence of AEs [130]. Notably, a remarkable fosfomycin bone/serum ratio (0.43 ± 0.04) was found in nine patients with diabetic foot infections receiving fosfomycin 100 mg/kg/day [131].
Several results documented the efficacy of cotrimoxazole, tetracyclines (i.e., doxycycline and minocycline), and clindamycin for suppressive prolonged therapy in PJIs, as also recommended by international guidelines [15]. In a retrospective cohort study including 78 patients affected by PJIs (72.1% Staphylococcus spp.) receiving long-term suppressive therapy with doxycycline (92%) or minocycline (8%), Pradier et al. [132] found a clinical failure rate of 28.2% after a mean follow-up of 1020 days. In three cases, a documented acquisition of tetracycline resistance in the index pathogen was documented, while AEs were reported in 18% of cases, of which 8% lead to tetracycline discontinuation [132]. In another retrospective study including 39 patients affected by PJIs (28.2% MRSA; 84.6% undergoing DAIR) receiving doxycycline-based suppressive antimicrobial therapy, clinical cure was reported in 74.4% of cases (20.5% relapse; 5.1% superinfections) [133]. AEs were reported in six patients (15.4%), leading to doxycycline discontinuation in two of them. In a multicenter retrospective study including 302 patients affected by PJIs (63.6% Staphylococcus spp.) receiving long-term suppressive antimicrobial therapy (tetracycline and cotrimoxazole in 39.7% and 35.4% of the cases, respectively) for a median time of 36.5 months, a clinical success rate of 58.6% was found. AEs leading to therapy discontinuation occurred in 5.6% of patients [134]. Twenty-three patients affected by PJIs after hip replacement and receiving long-term suppressive therapy (60.9% doxycycline; 26.1% cotrimoxazole) were retrospectively studied [135]. After a median follow-up of 33 months, clinical success was documented in 56.5% of patients, with relapse occurring in 7 cases. A total of 26.1% of patients experienced AEs during long-term suppressive antimicrobial therapy, leading to treatment discontinuation in two cases [135]. Twenty-one patients affected by PJIs mainly caused by Staphylococcus spp. and receiving suppressive antimicrobial therapy with minocycline (67%) or clindamycin (83%) were retrospectively assessed [136]. Clinical success was found in 67% of patients, while AEs requiring a switch of antibiotic treatment or dosage adjustment were reported in 43% of cases. Cotrimoxazole, mainly in association with fluoroquinolones or rifampicin, was found to be an effective salvage therapy in 51 patients affected by bone and joint infections (76.1% device-associated), with a favourable outcome in 78.4% of cases at 90 days [137]. Courjon et al. [138] retrospectively assessed 196 patients affected by bone and joint infections (41% device-associated; 81% Staphylococcus spp.) and treated with clindamycin (in combination therapy with fluoroquinolones or rifampicin in 31% and 27% of cases, respectively). After a mean follow-up of 28 months, clinical remission was found in 57% of patients (83% of those assessable), while AEs were recorded in only nine patients. Notably, no difference in terms of efficacy and safety was found in 18 patients receiving combination therapy with cotrimoxazole and rifampicin when compared to 18 cases treated with linezolid plus rifampicin as a prolonged oral therapy for orthopaedic implant-associated infections [88].
In vitro evidence showed poor antibiofilm activity of cotrimoxazole, clindamycin, fosfomycin, and eravacycline against S. aureus and S. epidermidis strains isolated from PJIs [83,139]. Interestingly, effective bone concentrations were found in different clinical studies for doxycycline, cotrimoxazole, and clindamycin, exceeding MIC50/90 of Staphylococcus isolates retrieved in orthopaedic implant-associated infections [7,120]. The bone/serum concentration ratio was 0.21–0.45 for clindamycin and 0.2–0.3 for cotrimoxazole [7,120].

3. Discussion

The choice of the best antibiotic strategy for the management of patients affected by orthopaedic implant-associated infections currently remains extremely challenging, requiring a careful assessment of the PK/PD features of the different alternatives. The ideal antibiotic should provide a well-documented efficacy in real-world experiences for the treatment of implant-associated infections, showing both good bone penetration and adequate anti-biofilm activity. Furthermore, considering that these infections commonly required a long duration of treatment, both safety/tolerability and feasibility for outpatient administration represent important issues (Figure 3).
Novel long-acting lipoglycopeptides (namely dalbavancin and oritavancin) showed optimal bone penetration and good anti-biofilm activity coupled with an excellent safety profile and long-term tolerability [10,140]. Real-world evidence demonstrated optimal efficacy of dalbavancin in this scenario, with an overall clinical cure rate exceeding 80%, thus making it a potential first-line choice for orthopaedic implant-associated infections. Although real-world evidence assessing the efficacy of oritavancin in implant-associated infections is currently limited, a promising role and a growing use could be supposed for this antibiotic. By virtue of their long half-life, both dalbavancin and oritavancin exhibit an ideal profile for the outpatient management of patients affected by implant-associated infections, considering that a single administration of 1200 mg for oritavancin, or a double administration of 1500 mg for dalbavancin one week apart is sufficient to achieve optimal concentrations for up to five weeks [140,141]. Therapeutic drug monitoring (TDM) may play a crucial role in providing real-time feedback for the estimated duration of the optimal treatment of staphylococcal orthopaedic infections with dalbavancin in each single patient [142].
As regard other novel anti-staphylococcal agents, real-world evidence assessing the efficacy of telavancin, ceftaroline, and ceftobiprole in orthopaedic implant-associated infections is still limited. Considering that these agents require a daily intravenous administration, their use in an outpatient regimen could be extremely challenging. Although telavancin, ceftaroline, and ceftobiprole may exhibit a promising anti-biofilm activity, further investigation is required. Additionally, both bone penetration and long-term safety of these agents remain an open issue.
The real-world use and the efficacy of daptomycin and linezolid in implant-associated infections is well-established. Both agents show excellent bone penetration, although their anti-biofilm activity is poor; thus, they should be used in combination therapy with rifampicin or other anti-biofilm agents. Considering its excellent oral bioavailability, linezolid is widely used as an outpatient therapeutic regimen for staphylococcal prosthetic infections. However, long-term safety represents a major issue, given that AEs (particularly myelotoxicity and neurotoxicity) occur in most of cases. TDM may prove to be remarkably helpful for improving safety outcomes in patients requiring long-term treatment with linezolid [143,144,145]. Although preliminary evidence showed a better safety profile and anti-biofilm activity of tedizolid compared to linezolid, further investigation is required [146]. Additionally, real-world evidence assessing the efficacy of tedizolid in prosthetic infections is very limited, while bone penetration remains an open issue. However, considering the physicochemical and PK features tedizolid, it could be suggested that no significant difference exists when compared to linezolid in terms of bone penetration.
Although vancomycin and teicoplanin are widely used for the management of orthopaedic prosthetic infections caused by MRSA, the lack of antibiofilm-activity coupled with poor bone penetration (particularly for vancomycin) represent major concerns. Furthermore, the need for daily intravenous administration makes vancomycin an unappealing alternative for outpatient management. Conversely, according to its long half-life, evidence demonstrates the feasibility of administering teicoplanin as an effective strategy for the outpatient management of patients affected by orthopaedic implant-associated infections [147,148].
Rifampicin currently represents a cornerstone in the management of staphylococcal orthopaedic implant-associated infections, considering its excellent anti-biofilm activity, good bone penetration, and feasibility for outpatient management. Rifampicin is commonly used as a companion drug for the management of PJIs caused by MSSA or MRSA managed with DAIR or one-stage exchange, as well as in a combination regimen with fluoroquinolones, tetracyclines, cotrimoxazole, or clindamycin for chronic oral antimicrobial suppression [15]. Long-term safety and tolerability represent a major concern for rifampicin and fluoroquinolones [149], and to a lesser degree, for cotrimoxazole and tetracyclines.
Although a longer follow-up duration was mainly implemented in studies assessing the role of daptomycin, linezolid, and fluoroquinolones when compared to novel lipoglycopeptides, this aspect had no impact on the feasibility of treatments, being essentially related to the different outcome definitions used in retrospective observational studies and case series.
Notably, while methods for assessing the MIC are well standardized, these are not fully established for determining the antibiofilm activity of a specific antibiotic. However, the methods and the criteria selected for the determination of the MBIC, MBBC, and MBEC values for the same agents are similar and comparable, thus having no impact on the assessment of the antibiofilm activity of the different antibiotics.
Local delivery of antibiotics may represent an attractive and complementary therapeutic strategy for the treatment of biofilm-associated orthopaedic implant-associated Staphylococcal infections [150,151]. In this scenario, the implant of antibiotic-loaded carrier material may ensure the achievement of high antibiotic concentrations locally, thus overcoming the poor vascularity and biofilm formation potentially involved in the failure of a systemic antibiotic approach [151]. Although polymethylmethacrylate (PMMA) was initially used as an antibiotic carrier, its undesirable features (the need for subsequent removal, potential biofilm formation on surface) caused it to be replaced by other absorbable materials (calcium sulphate beads, hydrogel) [151,152]. Vancomycin, gentamycin, and tobramycin are the most broadly used agents for local delivery [151,152,153]. The delivery of local rifampicin has raised concerns about the development of rapid resistance when used as a single agent [150]. Conversely, the addition of fosfomycin or dalbavancin to antibiotic-loaded bone cement in orthopaedic implant-associated Staphylococcal infections showed high concentrations, with promising data in experimental biofilm models [150,154]. Although the use of local antibiotic delivery seems to be promising, there is still a lack of level 1 evidence to demonstrate the efficacy of this strategy [150]. Currently, the evidence supports the implementation of local antibiotic treatments mainly in the prophylaxis of periprosthetic or fracture-related infections, while therapeutic use remains an unmet clinical need [152,155].
Considering the extreme challenge of the treatment of orthopaedic implant-associated infections, the implementation of a multidisciplinary taskforce managing each stage of these conditions is essential in order to improve clinical outcome. Specifically, clinical microbiologists play a crucial role in the diagnosis of orthopaedic prosthetic infections, providing the identification of causative pathogens and their susceptibility, thus allowing the implementation of the best targeted antibiotic therapy. In this scenario, the implementation and the development a “network lab,” in which a prompt and close collaboration between microbiologists and clinicians takes place, as performed in the Tuscany region, could represent a model of best practice in the management of implant-associated infections.
Limitations of our study have to be addressed. No systematic search was performed; thus, we cannot exclude that some articles are missing. However, the assessment of the predefined major determinants was adequately supported for each antibiotic. Most of the included studies were conducted retrospectively, with a limited sample size. Furthermore, thresholds for assessing the predefined major determinants were arbitrarily defined according to the specific long-standing experience and expertise of each single member. Finally, the methods selected for the determination of antibiofilm activity are not standardized as are those commonly used for MIC determination. However, the criteria used in the different studies are comparable, thus providing reliable results for each agent in terms of specific antibiofilm activity.

4. Materials and Methods

A literature search was conducted on PubMed-MEDLINE (from inception until 19 December 2021) in order to retrieve studies investigating the use of traditional and novel anti-staphylococcal agents in the management of infections associated with orthopaedic implants. All antibiotics with documented activity against methicillin-resistant Staphylococcus spp. and cited in international guidelines performed by the IDSA for the diagnosis and management of prosthetic joint infections [15] were included. Additionally, other agents with documented evidence for the management of infections associated with orthopaedic implants caused by methicillin-resistant Staphylococcus spp. (namely dalbavancin, oritavancin, telavancin, ceftaroline, ceftobiprole, tedizolid, and fosfomycin) were also included. Agents with activity only against methicillin-resistant Staphylococcus spp. and cited in the IDSA guidelines [15] were excluded.
A multidisciplinary task force composed of microbiologists, infectious disease specialists, orthopaedic surgeons, and intensive care physicians operating in the Tuscany region and characterized by specific expertise in the management of infections associated with orthopaedic implants identified different main topics focusing on unmet clinical needs in this scenario, namely diagnostic issues and the role of bacterial biofilm from a microbiological standpoint, and real-world evidence for the use of traditional and novel anti-staphylococcal agents, with a specific assessment of bone penetration, anti-biofilm activity, long-term safety, and feasibility for outpatient regimens. The definitive agreement for the identification and selection of each major determinant was reached by the multidisciplinary team after thorough discussion based on specific long-standing experiences and on the specific expertise of each single member in the management of infections associated with orthopaedic implants.
Specific thresholds were arbitrarily defined on the basis of the specific expertise of the different members in order to assess the performance of selected antibiotics for each of the five major determinants:
(a)
Efficacy in implant-associated infections: at least three studies including more than 70 patients and an overall positive clinical outcome of at least 70% for scoring the performance of the agent as optimal;
(b)
Bone penetration: existence of at least one preclinical/clinical study reporting the achievement of antibiotic bone concentrations above the MIC50/MIC90 for Staphylococcus spp. for scoring the performance of the agent as optimal;
(c)
Antibiofilm activity: existence of at least one in vitro study reporting the potential achievement of antibiotic concentrations above the MBIC for scoring the performance of the agent as optimal;
(d)
Long-term safety: an overall AEs rate below 20% in the included clinical studies for scoring the performance of the agent as optimal, while a proportion of the overall AEs above 40% identified agents with poor long-term safety;
(e)
Feasibility for outpatient management: the availability of oral formulations, and/or the possibility to perform once/twice-weekly administration, as well as feasibility for outpatient parenteral antibiotic therapy were considered for scoring the performance of the agent as optimal.
The following terms were searched on PubMed in combination: prosthetic joint infections, orthopaedic implant infections, bone infections, biofilm, bacterial biofilm, bone penetration, long-term, safety, dalbavancin, oritavancin, telavancin, daptomycin, linezolid, tedizolid, teicoplanin, ceftaroline, ceftobiprole, rifampicin, levofloxacin, fosfomycin, cotrimoxazole, minocycline, doxycycline, and tetracycline.
For each included study, the following information was extracted: (a) study author and year of publication; (b) study characteristics, including study design and sample size; (c) features of the patients, including site of infection, isolated pathogens, antibiotic therapy and duration, dosing schedule, and duration of follow-up; (d) types of outcome measurements, including rate of clinical success or improvement, clinical and/or microbiological failure rate, mortality rate, relapse rate, resistance development, and the overall proportion of adverse events.
The quality of the evidence was established according to a hierarchical scale of the study design, as reported in the evidence pyramid [156]: randomized controlled trials (RCTs); prospective observational studies; retrospective observational studies; case series; case reports; preclinical/in vitro studies. Clinical and preclinical pharmacokinetic or pharmacokinetic/pharmacodynamic (PK/PD) studies, as well as microbiological studies, were retrieved in order to support the choice of each selected anti-staphylococcal agent.
The consistency between the retrieved studies was also considered by assessing the concordance in clinical outcome (for clinical studies) and/or specific endpoints (for preclinical/in vitro studies) of the included studies at each level of the evidence pyramid. Only articles published in English were included, and the search was focused mainly on the last ten years in order to provide an up-to-date overview on the scientific evidence that may support the therapeutic choice.

5. Conclusions

The implementation of the best antibiotic strategy in patients affected by orthopaedic implant-associated infections remains extremely challenging, persisting as a matter of debate despite available guidelines. In this scenario, the existence of firm evidence establishing the efficacy of the different agents, stemming from well-conducted and possibly comparative studies, is crucial for guiding physicians in the choice of the best therapeutic strategy. Furthermore, the assessment of adequate bone penetration and anti-biofilm activity, coupled with long-term tolerability and feasibility for outpatient management, represent key issues for the treatment of orthopaedic implant-associated infections. Among available antibiotic treatments, novel long-acting lipoglycopeptide, particularly dalbavancin, alone or in combination with rifampicin, could represent the best antibiotic choice according to real-world evidence and PK/PD properties. The implementation of a multidisciplinary taskforce and of a close cooperation between microbiologists and clinicians is crucial for providing the best care in this scenario. The clinical interpretation of the TDM of the different antibiotics performed by a well-trained clinical pharmacologist could represent an added value for maximizing the clinical efficacy and minimizing toxicity in patients affected by orthopaedic implant-associated infections.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antibiotics11030406/s1; Table S1: A summary of the evidence investigating the off-label use of novel lipoglycopeptides for the management of infections associated with orthopaedic implants; Table S2: A summary of the evidence investigating the off-label use of novel anti-staphylococcal cephalosporins for the management of infections associated with orthopaedic implants; Table S3: A summary of the evidence investigating the off-label use of daptomycin for the management of infections associated with orthopaedic implants; Table S4: A summary of the evidence investigating the off-label use of linezolid and tedizolid for the management of infections associated with orthopaedic implants; Table S5: A summary of the evidence investigating the off-label use of fluoroquinolones for the management of infections associated with orthopaedic implants.

Author Contributions

Conceptualization, M.G., S.B., F.G. (Fabio Guarracino), E.M.P., M.S., B.V., S.D., S.F., A.G. and F.G. (Fabrizio Gemmi); methodology, M.G.; data curation, M.G.; writing—original draft preparation, M.G.; writing—review and editing, S.B., F.G. (Fabio Guarracino), E.M.P., M.S., B.V., S.D., S.F., A.G. and F.G. (Fabrizio Gemmi). All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by an unrestricted grant from Angelini S.p.A.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are retrieved from publicly available papers.

Conflicts of Interest

M.G. received personal fees from Angelini; B.V. participated on advisory boards, in a speaker’s bureau, and received research contracts, contributions and study grants from Abbott, Accelerate Diagnostics, Ada, Alifax, Angelini, Becton, Dickinson and Company, Bellco Drug Corporation, bioMerieux, Inc., Biotest, Cepheid, Correvio, Gilead, Menarini, MSD Italia, Nordic Pharma, Pfizer, Shionogi Inc., and Thermo Fisher Scientific; A.G. received personal fees from Menarini and Medivis.

References

  1. Trampuz, A.; Widmer, A.F. Infections Associated with Orthopedic Implants. Curr. Opin. Infect. Dis. 2006, 19, 349–356. [Google Scholar] [CrossRef] [PubMed]
  2. Tande, A.J.; Patel, R. Prosthetic Joint Infection. Clin. Microbiol. Rev. 2014, 27, 302–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zimmerli, W. Clinical Presentation and Treatment of Orthopaedic Implant-Associated Infection. J. Intern. Med. 2014, 276, 111–119. [Google Scholar] [CrossRef] [PubMed]
  4. Zimmerli, W.; Trampuz, A.; Ochsner, P.E. Prosthetic-Joint Infections. N. Engl. J. Med. 2004, 351, 1645–1654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Peel, T.N.; Cheng, A.C.; Buising, K.L.; Choong, P.F.M. Microbiological Aetiology, Epidemiology, and Clinical Profile of Prosthetic Joint Infections: Are Current Antibiotic Prophylaxis Guidelines Effective? Antimicrob. Agents Chemother. 2012, 56, 2386–2391. [Google Scholar] [CrossRef] [Green Version]
  6. Pea, F.; Viale, P. Bench-to-Bedside Review: Appropriate Antibiotic Therapy in Severe Sepsis and Septic Shock—Does the Dose Matter? Crit. Care 2009, 13, 214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Thabit, A.K.; Fatani, D.F.; Bamakhrama, M.S.; Barnawi, O.A.; Basudan, L.O.; Alhejaili, S.F. Antibiotic Penetration into Bone and Joints: An Updated Review. Int. J. Infect. Dis. 2019, 81, 128–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Pea, F. Penetration of Antibacterials into Bone: What Do We Really Need to Know for Optimal Prophylaxis and Treatment of Bone and Joint Infections? Clin. Pharm. 2009, 48, 125–127. [Google Scholar] [CrossRef]
  9. Jacqueline, C.; Caillon, J. Impact of Bacterial Biofilm on the Treatment of Prosthetic Joint Infections. J. Antimicrob. Chemother. 2014, 69 (Suppl. S1), i37–i40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Gatti, M.; Andreoni, M.; Pea, F.; Viale, P. Real-World Use of Dalbavancin in the Era of Empowerment of Outpatient Antimicrobial Treatment: A Careful Appraisal beyond Approved Indications Focusing on Unmet Clinical Needs. Drug Des. Dev. Ther. 2021, 15, 3349–3378. [Google Scholar] [CrossRef] [PubMed]
  11. Viale, P.; Tedeschi, S.; Scudeller, L.; Attard, L.; Badia, L.; Bartoletti, M.; Cascavilla, A.; Cristini, F.; Dentale, N.; Fasulo, G.; et al. Infectious Diseases Team for the Early Management of Severe Sepsis and Septic Shock in the Emergency Department. Clin. Infect. Dis. 2017, 65, 1253–1259. [Google Scholar] [CrossRef] [PubMed]
  12. Gatti, M.; Gasparini, L.E.; Laratta, M.; Sigurtà, A.; Rossi, A.; Brioschi, P.; Chiara, O.; Vismara, C.; Scaglione, F.; Arlati, S. Intensive Multidisciplinary Management in Critical Care Patients Affected by Severe Necrotizing Soft Tissue Infections: A Cooperative Method to Improve the Efficacy of Treatment. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 1153–1162. [Google Scholar] [CrossRef] [PubMed]
  13. Approvazione Linee Di Indirizzo per Un Approccio Integrato Alla Prevenzione e Contrasto Alle Infezioni Correlate All’assistenza, All’antimicrobico-Resistenza e Alla Sepsi—Regione Toscana—Delibera n.1439 Del 17-12-2018 2018. Available online: http://www301.regione.toscana.it/bancadati/atti/DettaglioAttiG.xml?codprat=2018DG00000001574 (accessed on 10 January 2022).
  14. L’antibiotico-Resistenza E L’uso Di Antibiotici in Toscana. 2021. Available online: https://www.ars.toscana.it/images/pubblicazioni/Collana_ARS/2020/Documento_ARS_107/Doc_112_SMART_2021_DEF.pdf (accessed on 10 January 2022).
  15. Osmon, D.R.; Berbari, E.F.; Berendt, A.R.; Lew, D.; Zimmerli, W.; Steckelberg, J.M.; Rao, N.; Hanssen, A.; Wilson, W.R. Infectious Diseases Society of America Diagnosis and Management of Prosthetic Joint Infection: Clinical Practice Guidelines by the Infectious Diseases Society of America. Clin. Infect. Dis. 2013, 56, e1–e25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Anemüller, R.; Belden, K.; Brause, B.; Citak, M.; Del Pozo, J.L.; Frommelt, L.; Gehrke, T.; Hewlett, A.; Higuera, C.A.; Hughes, H.; et al. Hip and Knee Section, Treatment, Antimicrobials: Proceedings of International Consensus on Orthopedic Infections. J. Arthroplast. 2019, 34, S463–S475. [Google Scholar] [CrossRef]
  17. Minassian, A.M.; Osmon, D.R.; Berendt, A.R. Clinical Guidelines in the Management of Prosthetic Joint Infection. J. Antimicrob. Chemother. 2014, 69 (Suppl. S1), i29–i35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Atkins, B.L.; Athanasou, N.; Deeks, J.J.; Crook, D.W.; Simpson, H.; Peto, T.E.; McLardy-Smith, P.; Berendt, A.R. Prospective Evaluation of Criteria for Microbiological Diagnosis of Prosthetic-Joint Infection at Revision Arthroplasty. The OSIRIS Collaborative Study Group. J. Clin. Microbiol. 1998, 36, 2932–2939. [Google Scholar] [CrossRef] [Green Version]
  19. Trampuz, A.; Piper, K.E.; Jacobson, M.J.; Hanssen, A.D.; Unni, K.K.; Osmon, D.R.; Mandrekar, J.N.; Cockerill, F.R.; Steckelberg, J.M.; Greenleaf, J.F.; et al. Sonication of Removed Hip and Knee Prostheses for Diagnosis of Infection. N. Engl. J. Med. 2007, 357, 654–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Schäfer, P.; Fink, B.; Sandow, D.; Margull, A.; Berger, I.; Frommelt, L. Prolonged Bacterial Culture to Identify Late Periprosthetic Joint Infection: A Promising Strategy. Clin. Infect. Dis. 2008, 47, 1403–1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Roux, A.-L.; Sivadon-Tardy, V.; Bauer, T.; Lortat-Jacob, A.; Herrmann, J.-L.; Gaillard, J.-L.; Rottman, M. Diagnosis of Prosthetic Joint Infection by Beadmill Processing of a Periprosthetic Specimen. Clin. Microbiol. Infect. 2011, 17, 447–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Holinka, J.; Bauer, L.; Hirschl, A.M.; Graninger, W.; Windhager, R.; Presterl, E. Sonication Cultures of Explanted Components as an Add-on Test to Routinely Conducted Microbiological Diagnostics Improve Pathogen Detection. J. Orthop. Res. 2011, 29, 617–622. [Google Scholar] [CrossRef]
  23. Karbysheva, S.; Di Luca, M.; Butini, M.E.; Winkler, T.; Schütz, M.; Trampuz, A. Comparison of Sonication with Chemical Biofilm Dislodgement Methods Using Chelating and Reducing Agents: Implications for the Microbiological Diagnosis of Implant Associated Infection. PLoS ONE 2020, 15, e0231389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Suren, C.; Feihl, S.; Cabric, S.; Banke, I.J.; Haller, B.; Trampuz, A.; von Eisenhart-Rothe, R.; Prodinger, P.M. Improved Pre-Operative Diagnostic Accuracy for Low-Grade Prosthetic Joint Infections Using Second-Generation Multiplex Polymerase Chain Reaction on Joint Fluid Aspirate. Int. Orthop. 2020, 44, 1629–1637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Morgenstern, C.; Cabric, S.; Perka, C.; Trampuz, A.; Renz, N. Synovial Fluid Multiplex PCR Is Superior to Culture for Detection of Low-Virulent Pathogens Causing Periprosthetic Joint Infection. Diagn. Microbiol. Infect. Dis. 2018, 90, 115–119. [Google Scholar] [CrossRef] [PubMed]
  26. Kahl, B.C.; Becker, K.; Löffler, B. Clinical Significance and Pathogenesis of Staphylococcal Small Colony Variants in Persistent Infections. Clin. Microbiol. Rev. 2016, 29, 401–427. [Google Scholar] [CrossRef] [Green Version]
  27. Neut, D.; van der Mei, H.C.; Bulstra, S.K.; Busscher, H.J. The Role of Small-Colony Variants in Failure to Diagnose and Treat Biofilm Infections in Orthopedics. Acta Orthop. 2007, 78, 299–308. [Google Scholar] [CrossRef] [Green Version]
  28. Ciofu, O.; Rojo-Molinero, E.; Macià, M.D.; Oliver, A. Antibiotic Treatment of Biofilm Infections. APMIS 2017, 125, 304–319. [Google Scholar] [CrossRef]
  29. Morata, L.; Cobo, J.; Fernández-Sampedro, M.; Guisado Vasco, P.; Ruano, E.; Lora-Tamayo, J.; Sánchez Somolinos, M.; González Ruano, P.; Rico Nieto, A.; Arnaiz, A.; et al. Safety and Efficacy of Prolonged Use of Dalbavancin in Bone and Joint Infections. Antimicrob. Agents Chemother. 2019, 63, e02280-18. [Google Scholar] [CrossRef] [Green Version]
  30. Wunsch, S.; Krause, R.; Valentin, T.; Prattes, J.; Janata, O.; Lenger, A.; Bellmann-Weiler, R.; Weiss, G.; Zollner-Schwetz, I. Multicenter Clinical Experience of Real Life Dalbavancin Use in Gram-Positive Infections. Int. J. Infect. Dis. 2019, 81, 210–214. [Google Scholar] [CrossRef] [Green Version]
  31. Bai, F.; Aldieri, C.; Cattelan, A.; Raumer, F.; Di Meco, E.; Moioli, M.C.; Tordato, F.; Morelli, P.; Borghi, F.; Rizzi, M.; et al. Efficacy and Safety of Dalbavancin in the Treatment of Acute Bacterial Skin and Skin Structure Infections (ABSSSIs) and Other Infections in a Real-Life Setting: Data from an Italian Observational Multicentric Study (DALBITA Study). Expert Rev. Anti Infect. Ther. 2020, 18, 1271–1279. [Google Scholar] [CrossRef]
  32. Tobudic, S.; Forstner, C.; Burgmann, H.; Lagler, H.; Steininger, C.; Traby, L.; Vossen, M.G.; Winkler, S.; Thalhammer, F. Real-World Experience with Dalbavancin Therapy in Gram-Positive Skin and Soft Tissue Infection, Bone and Joint Infection. Infection 2019, 47, 1013–1020. [Google Scholar] [CrossRef] [Green Version]
  33. Bouza, E.; Valerio, M.; Soriano, A.; Morata, L.; Carus, E.G.; Rodríguez-González, C.; Hidalgo-Tenorio, M.C.; Plata, A.; Muñoz, P.; Vena, A. Dalbavancin in the Treatment of Different Gram-Positive Infections: A Real-Life Experience. Int. J. Antimicrob. Agents 2018, 51, 571–577. [Google Scholar] [CrossRef]
  34. Buzón Martín, L.; Mora Fernández, M.; Perales Ruiz, J.M.; Ortega Lafont, M.; Álvarez Paredes, L.; Morán Rodríguez, M.A.; Fernández Regueras, M.; Machín Morón, M.A.; Mejías Lobón, G. Dalbavancin for Treating Prosthetic Joint Infections Caused by Gram-Positive Bacteria: A Proposal for a Low Dose Strategy. A Retrospective Cohort Study. Rev. Esp. Quim. 2019, 32, 532–538. [Google Scholar]
  35. Bork, J.T.; Heil, E.L.; Berry, S.; Lopes, E.; Davé, R.; Gilliam, B.L.; Amoroso, A. Dalbavancin Use in Vulnerable Patients Receiving Outpatient Parenteral Antibiotic Therapy for Invasive Gram-Positive Infections. Infect. Dis. Ther. 2019, 8, 171–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Brescini, L.; Della Martera, F.; Morroni, G.; Mazzanti, S.; Di Pietrantonio, M.; Mantini, P.; Candelaresi, B.; Pallotta, F.; Olivieri, S.; Iencinella, V.; et al. Use of Dalbavancin in Skin, Bone and Joint Infections: A Real-Life Experience in an Italian Center. Antibiotics 2021, 10, 1129. [Google Scholar] [CrossRef] [PubMed]
  37. Núñez-Núñez, M.P.; Casas-Hidalgo, I.; García-Fumero, R.; Vallejo-Rodríguez, I.; Anguita-Santos, F.; Hernández-Quero, J.; Cabeza-Barrera, J.; Ruiz-Sancho, A. Dalbavancin Is a Novel Antimicrobial against Gram-Positive Pathogens: Clinical Experience beyond Labelled Indications. Eur. J. Hosp. Pharm. 2020, 27, 310–312. [Google Scholar] [CrossRef]
  38. Vazquez Deida, A.A.; Shihadeh, K.C.; Preslaski, C.R.; Young, H.L.; Wyles, D.L.; Jenkins, T.C. Use of a Standardized Dalbavancin Approach to Facilitate Earlier Hospital Discharge for Vulnerable Patients Receiving Prolonged Inpatient Antibiotic Therapy. Open Forum Infect. Dis. 2020, 7, ofaa293. [Google Scholar] [CrossRef]
  39. Azamgarhi, T.; Donaldson, J.; Shah, A.; Warren, S. Dalbavancin to Treat Infected Massive Endoprostheses: A Case Report and Cost Comparison Analysis. J. Bone Jt. Infect. 2019, 4, 234–237. [Google Scholar] [CrossRef]
  40. Trujillano Ruiz, A.; Mesquida Riera, J.; Serrano Fabiá, M.A.; Riera Pérez, E.; Mejía Benard, A.; Taberner Ferrer, M.D. Prolonged treatment with dalbavancin in prosthetic hip infection by methicillin-resistant Staphylococcus epidermidis. Rev. Esp. Quim. 2019, 32, 203–204. [Google Scholar]
  41. Carrión Madroñal, I.M.; Sánchez Del Moral, R.; Abad Zamora, J.M.; Martínez Marcos, F.J. Dalbavancin Combined with Linezolid in Prosthetic-Hip Infection. Rev. Esp. Quim. 2020, 33, 147–148. [Google Scholar] [CrossRef]
  42. Ramírez Hidalgo, M.; Jover-Sáenz, A.; García-González, M.; Barcenilla-Gaite, F. Dalbavancin Treatment of Prosthetic Knee Infection Due to Oxacillin-Resistant Staphylococcus epidermidis. Enferm. Infecc. Microbiol. Clin. 2018, 36, 142–143. [Google Scholar] [CrossRef]
  43. Fernández, J.; Greenwood-Quaintance, K.E.; Patel, R. In Vitro Activity of Dalbavancin against Biofilms of Staphylococci Isolated from Prosthetic Joint Infections. Diagn. Microbiol. Infect. Dis. 2016, 85, 449–451. [Google Scholar] [CrossRef] [PubMed]
  44. Schmidt-Malan, S.M.; Greenwood Quaintance, K.E.; Karau, M.J.; Patel, R. In Vitro Activity of Tedizolid against Staphylococci Isolated from Prosthetic Joint Infections. Diagn. Microbiol. Infect. Dis. 2016, 85, 77–79. [Google Scholar] [CrossRef]
  45. Knafl, D.; Tobudic, S.; Cheng, S.C.; Bellamy, D.R.; Thalhammer, F. Dalbavancin Reduces Biofilms of Methicillin-Resistant Staphylococcus aureus (MRSA) and Methicillin-Resistant Staphylococcus epidermidis (MRSE). Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 677–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Neudorfer, K.; Schmidt-Malan, S.M.; Patel, R. Dalbavancin Is Active in Vitro against Biofilms Formed by Dalbavancin-Susceptible Enterococci. Diagn. Microbiol. Infect. Dis. 2018, 90, 58–63. [Google Scholar] [CrossRef] [PubMed]
  47. Žiemytė, M.; Rodríguez-Díaz, J.C.; Ventero, M.P.; Mira, A.; Ferrer, M.D. Effect of Dalbavancin on Staphylococcal Biofilms When Administered Alone or in Combination With Biofilm-Detaching Compounds. Front. Microbiol. 2020, 11, 553. [Google Scholar] [CrossRef] [Green Version]
  48. Jacob, B.; Makarewicz, O.; Hartung, A.; Brodt, S.; Roehner, E.; Matziolis, G. In Vitro Additive Effects of Dalbavancin and Rifampicin against Biofilm of Staphylococcus aureus. Sci. Rep. 2021, 11, 23425. [Google Scholar] [CrossRef] [PubMed]
  49. Solon, E.G.; Dowell, J.A.; Lee, J.; King, S.P.; Damle, B.D. Distribution of Radioactivity in Bone and Related Structures Following Administration of [14C] Dalbavancin to New Zealand White Rabbits. Antimicrob. Agents Chemother. 2007, 51, 3008–3010. [Google Scholar] [CrossRef] [Green Version]
  50. Redell, M.; Sierra-Hoffman, M.; Assi, M.; Bochan, M.; Chansolme, D.; Gandhi, A.; Sheridan, K.; Soosaipillai, I.; Walsh, T.; Massey, J. The CHROME Study, a Real-World Experience of Single- and Multiple-Dose Oritavancin for Treatment of Gram-Positive Infections. Open Forum Infect. Dis. 2019, 6, ofz479. [Google Scholar] [CrossRef]
  51. Schulz, L.T.; Dworkin, E.; Dela-Pena, J.; Rose, W.E. Multiple-Dose Oritavancin Evaluation in a Retrospective Cohort of Patients with Complicated Infections. Pharmacotherapy 2018, 38, 152–159. [Google Scholar] [CrossRef]
  52. Yan, Q.; Karau, M.J.; Patel, R. In Vitro Activity of Oritavancin against Biofilms of Staphylococci Isolated from Prosthetic Joint Infection. Diagn. Microbiol. Infect. Dis. 2018, 92, 155–157. [Google Scholar] [CrossRef]
  53. Belley, A.; Neesham-Grenon, E.; McKay, G.; Arhin, F.F.; Harris, R.; Beveridge, T.; Parr, T.R.; Moeck, G. Oritavancin Kills Stationary-Phase and Biofilm Staphylococcus aureus Cells in Vitro. Antimicrob. Agents Chemother. 2009, 53, 918–925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Yan, Q.; Karau, M.J.; Raval, Y.S.; Patel, R. Evaluation of Oritavancin Combinations with Rifampin, Gentamicin, or Linezolid against Prosthetic Joint Infection-Associated Methicillin-Resistant Staphylococcus aureus Biofilms by Time-Kill Assays. Antimicrob. Agents Chemother. 2018, 62, e00943-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Lehoux, D.; Ostiguy, V.; Cadieux, C.; Malouin, M.; Belanger, O.; Far, A.R.; Parr, T.R. Oritavancin Pharmacokinetics and Bone Penetration in Rabbits. Antimicrob. Agents Chemother. 2015, 59, 6501–6505. [Google Scholar] [CrossRef] [Green Version]
  56. Sims, C.R.; Bressler, A.M.; Graham, D.R.; Lacy, M.K.; Lombardi, D.A.; Castaneda-Ruiz, B. Real-World Clinical Use and Outcomes of Telavancin for the Treatment of Bone and Joint Infections: Results from the Telavancin Observational Use Registry (TOURTM). Drugs Real World Outcomes 2021, 8, 509–518. [Google Scholar] [CrossRef]
  57. Harting, J.; Fernandez, F.; Kelley, R.; Wiemken, T.; Peyrani, P.; Ramirez, J. Telavancin for the Treatment of Methicillin-Resistant Staphylococcus aureus Bone and Joint Infections. Diagn. Microbiol. Infect. Dis. 2017, 89, 294–299. [Google Scholar] [CrossRef]
  58. LaPlante, K.L.; Mermel, L.A. In Vitro Activities of Telavancin and Vancomycin against Biofilm-Producing Staphylococcus aureus, S. epidermidis, and Enterococcus faecalis Strains. Antimicrob. Agents Chemother. 2009, 53, 3166–3169. [Google Scholar] [CrossRef] [Green Version]
  59. Buzón-Martín, L.; Zollner-Schwetz, I.; Tobudic, S.; Cercenado, E.; Lora-Tamayo, J. Dalbavancin for the Treatment of Prosthetic Joint Infections: A Narrative Review. Antibiotics 2021, 10, 656. [Google Scholar] [CrossRef]
  60. Zhanel, G.G.; Calic, D.; Schweizer, F.; Zelenitsky, S.; Adam, H.; Lagacé-Wiens, P.R.S.; Rubinstein, E.; Gin, A.S.; Hoban, D.J.; Karlowsky, J.A. New Lipoglycopeptides: A Comparative Review of Dalbavancin, Oritavancin and Telavancin. Drugs 2010, 70, 859–886. [Google Scholar] [CrossRef]
  61. Malandain, D.; Dinh, A.; Ferry, T.; Touchais, S.; Lustig, S.; Laurent, F.; Corvec, S.; Bémer, P.; Asseray, N.; Boutoille, D. Salvage Therapy for Complex Bone and Joint Infections with Ceftaroline: A Multicentre, Observational Study. Int. J. Antimicrob. Agents 2017, 50, 277–280. [Google Scholar] [CrossRef] [PubMed]
  62. Park, K.-H.; Greenwood-Quaintance, K.E.; Patel, R. In Vitro Activity of Ceftaroline against Staphylococci from Prosthetic Joint Infection. Diagn. Microbiol. Infect. Dis. 2016, 84, 141–143. [Google Scholar] [CrossRef] [PubMed]
  63. Landini, G.; Riccobono, E.; Giani, T.; Arena, F.; Rossolini, G.M.; Pallecchi, L. Bactericidal Activity of Ceftaroline against Mature Staphylococcus aureus Biofilms. Int. J. Antimicrob. Agents 2015, 45, 551–553. [Google Scholar] [CrossRef] [PubMed]
  64. Barber, K.E.; Smith, J.R.; Ireland, C.E.; Boles, B.R.; Rose, W.E.; Rybak, M.J. Evaluation of Ceftaroline Alone and in Combination against Biofilm-Producing Methicillin-Resistant Staphylococcus aureus with Reduced Susceptibility to Daptomycin and Vancomycin in an In Vitro Pharmacokinetic/Pharmacodynamic Model. Antimicrob. Agents Chemother. 2015, 59, 4497–4503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Gatin, L.; Saleh-Mghir, A.; Tasse, J.; Ghout, I.; Laurent, F.; Crémieux, A.-C. Ceftaroline-Fosamil Efficacy against Methicillin-Resistant Staphylococcus aureus in a Rabbit Prosthetic Joint Infection Model. Antimicrob. Agents Chemother. 2014, 58, 6496–6500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zhanel, G.G.; Kosar, J.; Baxter, M.; Dhami, R.; Borgia, S.; Irfan, N.; MacDonald, K.S.; Dow, G.; Lagacé-Wiens, P.; Dube, M.; et al. Real-Life Experience with Ceftobiprole in Canada: Results from the CLEAR (CanadianLEadership OnAntimicrobialReal-Life Usage) Registry. J. Glob. Antimicrob. Resist. 2021, 24, 335–339. [Google Scholar] [CrossRef]
  67. Abbanat, D.; Shang, W.; Amsler, K.; Santoro, C.; Baum, E.; Crespo-Carbone, S.; Lynch, A.S. Evaluation of the in Vitro Activities of Ceftobiprole and Comparators in Staphylococcal Colony or Microtitre Plate Biofilm Assays. Int. J. Antimicrob. Agents 2014, 43, 32–39. [Google Scholar] [CrossRef] [PubMed]
  68. Yin, L.-Y.; Calhoun, J.H.; Thomas, J.K.; Shapiro, S.; Schmitt-Hoffmann, A. Efficacies of Ceftobiprole Medocaril and Comparators in a Rabbit Model of Osteomyelitis Due to Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2008, 52, 1618–1622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Byren, I.; Rege, S.; Campanaro, E.; Yankelev, S.; Anastasiou, D.; Kuropatkin, G.; Evans, R. Randomized Controlled Trial of the Safety and Efficacy of Daptomycin versus Standard-of-Care Therapy for Management of Patients with Osteomyelitis Associated with Prosthetic Devices Undergoing Two-Stage Revision Arthroplasty. Antimicrob. Agents Chemother. 2012, 56, 5626–5632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Carli, A.V.; Miller, A.O.; Kapadia, M.; Chiu, Y.-F.; Westrich, G.H.; Brause, B.D.; Henry, M.W. Assessing the Role of Daptomycin as Antibiotic Therapy for Staphylococcal Prosthetic Joint Infection. J. Bone Jt. Infect. 2020, 5, 82–88. [Google Scholar] [CrossRef] [Green Version]
  71. Joseph, C.; Robineau, O.; Titecat, M.; Putman, S.; Blondiaux, N.; Loiez, C.; Valette, M.; Schmit, J.L.; Beltrand, E.; Dézeque, H.; et al. Daptomycin versus Vancomycin as Post-Operative Empirical Antibiotic Treatment for Prosthetic Joint Infections: A Case-Control Study. J. Bone Jt. Infect. 2019, 4, 72–75. [Google Scholar] [CrossRef] [Green Version]
  72. Rao, N.; Regalla, D.M. Uncertain Efficacy of Daptomycin for Prosthetic Joint Infections: A Prospective Case Series. Clin. Orthop. Relat. Res. 2006, 451, 34–37. [Google Scholar] [CrossRef] [PubMed]
  73. Kuo, F.-C.; Yen, S.-H.; Peng, K.-T.; Wang, J.-W.; Lee, M.S. Methicillin-Resistant Staphylococcal Periprosthetic Joint Infections Can Be Effectively Controlled by Systemic and Local Daptomycin. BMC Infect. Dis. 2016, 16, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Herrera, S.; Sorlí, L.; Horcajada, J.P. High-Dose Daptomycin Together with Rifampin as Salvage Therapy for Prosthetic Joint Infections. Med. Clin. (Barc.) 2017, 149, 223–224. [Google Scholar] [CrossRef]
  75. Corona Pérez-Cardona, P.S.; Barro Ojeda, V.; Rodriguez Pardo, D.; Pigrau Serrallach, C.; Guerra Farfán, E.; Amat Mateu, C.; Flores Sanchez, X. Clinical Experience with Daptomycin for the Treatment of Patients with Knee and Hip Periprosthetic Joint Infections. J. Antimicrob. Chemother. 2012, 67, 1749–1754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Lora-Tamayo, J.; Parra-Ruiz, J.; Rodríguez-Pardo, D.; Barberán, J.; Ribera, A.; Tornero, E.; Pigrau, C.; Mensa, J.; Ariza, J.; Soriano, A. High Doses of Daptomycin (10 Mg/Kg/d) plus Rifampin for the Treatment of Staphylococcal Prosthetic Joint Infection Managed with Implant Retention: A Comparative Study. Diagn. Microbiol. Infect. Dis. 2014, 80, 66–71. [Google Scholar] [CrossRef]
  77. Chang, Y.-J.; Lee, M.S.; Lee, C.-H.; Lin, P.-C.; Kuo, F.-C. Daptomycin Treatment in Patients with Resistant Staphylococcal Periprosthetic Joint Infection. BMC Infect. Dis. 2017, 17, 736. [Google Scholar] [CrossRef]
  78. Yuste, J.R.; Quesada, M.; Díaz-Rada, P.; Del Pozo, J.L. Daptomycin in the Treatment of Prosthetic Joint Infection by Enterococcus faecalis: Safety and Efficacy of High-Dose and Prolonged Therapy. Int. J. Infect. Dis. 2014, 27, 65–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Luengo, G.; Lora-Tamayo, J.; Paredes, D.; Muñoz-Gallego, I.; Díaz, A.; Delgado, E. Daptomycin Plus Fosfomycin as Salvage Therapy in a Difficult-to-Treat Total Femoral Replacement Infection. J. Bone Jt. Infect. 2018, 3, 207–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Montange, D.; Berthier, F.; Leclerc, G.; Serre, A.; Jeunet, L.; Berard, M.; Muret, P.; Vettoretti, L.; Leroy, J.; Hoen, B.; et al. Penetration of Daptomycin into Bone and Synovial Fluid in Joint Replacement. Antimicrob. Agents Chemother. 2014, 58, 3991–3996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Molina-Manso, D.; del Prado, G.; Ortiz-Pérez, A.; Manrubia-Cobo, M.; Gómez-Barrena, E.; Cordero-Ampuero, J.; Esteban, J. In Vitro Susceptibility to Antibiotics of Staphylococci in Biofilms Isolated from Orthopaedic Infections. Int. J. Antimicrob. Agents 2013, 41, 521–523. [Google Scholar] [CrossRef] [PubMed]
  82. Garrigós, C.; Murillo, O.; Euba, G.; Verdaguer, R.; Tubau, F.; Cabellos, C.; Cabo, J.; Ariza, J. Efficacy of Usual and High Doses of Daptomycin in Combination with Rifampin versus Alternative Therapies in Experimental Foreign-Body Infection by Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2010, 54, 5251–5256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Jahanbakhsh, S.; Singh, N.B.; Yim, J.; Kebriaei, R.; Smith, J.R.; Lev, K.; Tran, T.T.; Rose, W.E.; Arias, C.A.; Rybak, M.J. Impact of Daptomycin Dose Exposure Alone or in Combination with β-Lactams or Rifampin against Vancomycin-Resistant Enterococci in an In Vitro Biofilm Model. Antimicrob. Agents Chemother. 2020, 64, e02074-19. [Google Scholar] [CrossRef] [PubMed]
  84. Soriano, A.; Gómez, J.; Gómez, L.; Azanza, J.R.; Pérez, R.; Romero, F.; Pons, M.; Bella, F.; Velasco, M.; Mensa, J. Efficacy and Tolerability of Prolonged Linezolid Therapy in the Treatment of Orthopedic Implant Infections. Eur. J. Clin. Microbiol. Infect. Dis. 2007, 26, 353–356. [Google Scholar] [CrossRef] [PubMed]
  85. Gómez, J.; Canovas, E.; Baños, V.; Martínez, L.; García, E.; Hernández-Torres, A.; Canteras, M.; Ruiz, J.; Medina, M.; Martínez, P.; et al. Linezolid plus Rifampin as a Salvage Therapy in Prosthetic Joint Infections Treated without Removing the Implant. Antimicrob. Agents Chemother. 2011, 55, 4308–4310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Rao, N.; Hamilton, C.W. Efficacy and Safety of Linezolid for Gram-Positive Orthopedic Infections: A Prospective Case Series. Diagn. Microbiol. Infect. Dis. 2007, 59, 173–179. [Google Scholar] [CrossRef] [PubMed]
  87. Cobo, J.; Lora-Tamayo, J.; Euba, G.; Jover-Sáenz, A.; Palomino, J.; del Toro, M.D.; Rodríguez-Pardo, D.; Riera, M.; Ariza, J.; Red Española para la Investigación en Patología Infecciosa (REIPI). Linezolid in Late-Chronic Prosthetic Joint Infection Caused by Gram-Positive Bacteria. Diagn. Microbiol. Infect. Dis. 2013, 76, 93–98. [Google Scholar] [CrossRef] [PubMed]
  88. Nguyen, S.; Pasquet, A.; Legout, L.; Beltrand, E.; Dubreuil, L.; Migaud, H.; Yazdanpanah, Y.; Senneville, E. Efficacy and Tolerance of Rifampicin-Linezolid Compared with Rifampicin-Cotrimoxazole Combinations in Prolonged Oral Therapy for Bone and Joint Infections. Clin. Microbiol. Infect. 2009, 15, 1163–1169. [Google Scholar] [CrossRef] [Green Version]
  89. Papadopoulos, A.; Plachouras, D.; Giannitsioti, E.; Poulakou, G.; Giamarellou, H.; Kanellakopoulou, K. Efficacy and Tolerability of Linezolid in Chronic Osteomyelitis and Prosthetic Joint Infections: A Case-Control Study. J. Chemother. 2009, 21, 165–169. [Google Scholar] [CrossRef] [PubMed]
  90. Morata, L.; Senneville, E.; Bernard, L.; Nguyen, S.; Buzelé, R.; Druon, J.; Tornero, E.; Mensa, J.; Soriano, A. A Retrospective Review of the Clinical Experience of Linezolid with or Without Rifampicin in Prosthetic Joint Infections Treated with Debridement and Implant Retention. Infect. Dis. Ther. 2014, 3, 235–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Senneville, E.; Legout, L.; Valette, M.; Yazdanpanah, Y.; Beltrand, E.; Caillaux, M.; Migaud, H.; Mouton, Y. Effectiveness and Tolerability of Prolonged Linezolid Treatment for Chronic Osteomyelitis: A Retrospective Study. Clin. Ther. 2006, 28, 1155–1163. [Google Scholar] [CrossRef]
  92. Bassetti, M.; Vitale, F.; Melica, G.; Righi, E.; Di Biagio, A.; Molfetta, L.; Pipino, F.; Cruciani, M.; Bassetti, D. Linezolid in the Treatment of Gram-Positive Prosthetic Joint Infections. J. Antimicrob. Chemother. 2005, 55, 387–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Razonable, R.R.; Osmon, D.R.; Steckelberg, J.M. Linezolid Therapy for Orthopedic Infections. Mayo Clin. Proc. 2004, 79, 1137–1144. [Google Scholar] [CrossRef]
  94. Vercillo, M.; Patzakis, M.J.; Holtom, P.; Zalavras, C.G. Linezolid in the Treatment of Implant-Related Chronic Osteomyelitis. Clin. Orthop. Relat. Res. 2007, 461, 40–43. [Google Scholar] [CrossRef] [PubMed]
  95. Oussedik, S.I.S.; Haddad, F.S. The Use of Linezolid in the Treatment of Infected Total Joint Arthroplasty. J. Arthroplast. 2008, 23, 273–278. [Google Scholar] [CrossRef]
  96. Benavent, E.; Morata, L.; Escrihuela-Vidal, F.; Reynaga, E.A.; Soldevila, L.; Albiach, L.; Pedro-Botet, M.L.; Padullés, A.; Soriano, A.; Murillo, O. Long-Term Use of Tedizolid in Osteoarticular Infections: Benefits among Oxazolidinone Drugs. Antibiotics 2021, 10, 53. [Google Scholar] [CrossRef] [PubMed]
  97. Ferry, T.; Batailler, C.; Conrad, A.; Triffault-Fillit, C.; Laurent, F.; Valour, F.; Chidiac, C.; Lyon BJI Study Group. Correction of Linezolid-Induced Myelotoxicity After Switch to Tedizolid in a Patient Requiring Suppressive Antimicrobial Therapy for Multidrug-Resistant Staphylococcus epidermidis Prosthetic-Joint Infection. Open Forum Infect. Dis. 2018, 5, ofy246. [Google Scholar] [CrossRef] [PubMed]
  98. Rana, B.; Butcher, I.; Grigoris, P.; Murnaghan, C.; Seaton, R.A.; Tobin, C.M. Linezolid Penetration into Osteo-Articular Tissues. J. Antimicrob. Chemother. 2002, 50, 747–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Kutscha-Lissberg, F.; Hebler, U.; Muhr, G.; Köller, M. Linezolid Penetration into Bone and Joint Tissues Infected with Methicillin-Resistant Staphylococci. Antimicrob. Agents Chemother. 2003, 47, 3964–3966. [Google Scholar] [CrossRef] [Green Version]
  100. Abad, L.; Tafani, V.; Tasse, J.; Josse, J.; Chidiac, C.; Lustig, S.; Ferry, T.; Diot, A.; Laurent, F.; Valour, F. Evaluation of the Ability of Linezolid and Tedizolid to Eradicate Intraosteoblastic and Biofilm-Embedded Staphylococcus aureus in the Bone and Joint Infection Setting. J. Antimicrob. Chemother. 2019, 74, 625–632. [Google Scholar] [CrossRef] [PubMed]
  101. El Haj, C.; Murillo, O.; Ribera, A.; Lloberas, N.; Gómez-Junyent, J.; Tubau, F.; Fontova, P.; Cabellos, C.; Ariza, J. Evaluation of Linezolid or Trimethoprim/Sulfamethoxazole in Combination with Rifampicin as Alternative Oral Treatments Based on an in Vitro Pharmacodynamic Model of Staphylococcal Biofilm. Int. J. Antimicrob. Agents 2018, 51, 854–861. [Google Scholar] [CrossRef] [PubMed]
  102. Rappo, U.; Puttagunta, S.; Shevchenko, V.; Shevchenko, A.; Jandourek, A.; Gonzalez, P.L.; Suen, A.; Mas Casullo, V.; Melnick, D.; Miceli, R.; et al. Dalbavancin for the Treatment of Osteomyelitis in Adult Patients: A Randomized Clinical Trial of Efficacy and Safety. Open Forum Infect. Dis. 2019, 6, ofy331. [Google Scholar] [CrossRef] [PubMed]
  103. Veve, M.P.; Patel, N.; Smith, Z.A.; Yeager, S.D.; Wright, L.R.; Shorman, M.A. Comparison of Dalbavancin to Standard-of-Care for Outpatient Treatment of Invasive Gram-Positive Infections. Int. J. Antimicrob. Agents 2020, 56, 106210. [Google Scholar] [CrossRef] [PubMed]
  104. Graziani, A.L.; Lawson, L.A.; Gibson, G.A.; Steinberg, M.A.; MacGregor, R.R. Vancomycin Concentrations in Infected and Noninfected Human Bone. Antimicrob. Agents Chemother. 1988, 32, 1320–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Bue, M.; Tøttrup, M.; Hanberg, P.; Langhoff, O.; Birke-Sørensen, H.; Thillemann, T.M.; Andersson, T.L.; Søballe, K. Bone and Subcutaneous Adipose Tissue Pharmacokinetics of Vancomycin in Total Knee Replacement Patients. Acta Orthop. 2018, 89, 95–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Barberán, J. Management of Infections of Osteoarticular Prosthesis. Clin. Microbiol. Infect. 2006, 12 (Suppl. S3), 93–101. [Google Scholar] [CrossRef] [Green Version]
  107. Peeters, O.; Ferry, T.; Ader, F.; Boibieux, A.; Braun, E.; Bouaziz, A.; Karsenty, J.; Forestier, E.; Laurent, F.; Lustig, S.; et al. Teicoplanin-Based Antimicrobial Therapy in Staphylococcus aureus Bone and Joint Infection: Tolerance, Efficacy and Experience with Subcutaneous Administration. BMC Infect. Dis. 2016, 16, 622. [Google Scholar] [CrossRef] [Green Version]
  108. Pavoni, G.L.; Giannella, M.; Falcone, M.; Scorzolini, L.; Liberatore, M.; Carlesimo, B.; Serra, P.; Venditti, M. Conservative Medical Therapy of Prosthetic Joint Infections: Retrospective Analysis of an 8-Year Experience. Clin. Microbiol. Infect. 2004, 10, 831–837. [Google Scholar] [CrossRef] [Green Version]
  109. Claessens, J.; Roriz, M.; Merckx, R.; Baatsen, P.; Van Mellaert, L.; Van Eldere, J. Inefficacy of Vancomycin and Teicoplanin in Eradicating and Killing Staphylococcus epidermidis Biofilms in Vitro. Int. J. Antimicrob. Agents 2015, 45, 368–375. [Google Scholar] [CrossRef]
  110. Wilson, A.P.; Taylor, B.; Treasure, T.; Grüneberg, R.N.; Patton, K.; Felmingham, D.; Sturridge, M.F. Antibiotic Prophylaxis in Cardiac Surgery: Serum and Tissue Levels of Teicoplanin, Flucloxacillin and Tobramycin. J. Antimicrob. Chemother. 1988, 21, 201–212. [Google Scholar] [CrossRef]
  111. Nehrer, S.; Thalhammer, F.; Schwameis, E.; Breyer, S.; Kotz, R. Teicoplanin in the Prevention of Infection in Total Hip Replacement. Arch. Orthop. Trauma. Surg. 1998, 118, 32–36. [Google Scholar] [CrossRef]
  112. de Lalla, F.; Novelli, A.; Pellizzer, G.; Milocchi, F.; Viola, R.; Rigon, A.; Stecca, C.; Dal Pizzol, V.; Fallani, S.; Periti, P. Regional and Systemic Prophylaxis with Teicoplanin in Monolateral and Bilateral Total Knee Replacement Procedures: Study of Pharmacokinetics and Tissue Penetration. Antimicrob. Agents Chemother. 1993, 37, 2693–2698. [Google Scholar] [CrossRef] [Green Version]
  113. Lazzarini, L.; Novelli, A.; Marzano, N.; Timillero, L.; Fallani, S.; Viola, R.; de Lalla, F. Regional and Systemic Prophylaxis with Teicoplanin in Total Knee Arthroplasty: A Tissue Penetration Study. J. Arthroplast. 2003, 18, 342–346. [Google Scholar] [CrossRef] [PubMed]
  114. Zimmerli, W.; Sendi, P. Role of Rifampin against Staphylococcal Biofilm Infections In Vitro, in Animal Models, and in Orthopedic-Device-Related Infections. Antimicrob. Agents Chemother. 2019, 63, e01746-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Renz, N.; Trampuz, A.; Zimmerli, W. Controversy about the Role of Rifampin in Biofilm Infections: Is It Justified? Antibiotics 2021, 10, 165. [Google Scholar] [CrossRef] [PubMed]
  116. Zimmerli, W.; Widmer, A.F.; Blatter, M.; Frei, R.; Ochsner, P.E. Role of Rifampin for Treatment of Orthopedic Implant-Related Staphylococcal Infections: A Randomized Controlled Trial. Foreign-Body Infection (FBI) Study Group. JAMA 1998, 279, 1537–1541. [Google Scholar] [CrossRef] [Green Version]
  117. Karlsen, Ø.E.; Borgen, P.; Bragnes, B.; Figved, W.; Grøgaard, B.; Rydinge, J.; Sandberg, L.; Snorrason, F.; Wangen, H.; Witsøe, E.; et al. Rifampin Combination Therapy in Staphylococcal Prosthetic Joint Infections: A Randomized Controlled Trial. J. Orthop. Surg. Res. 2020, 15, 365. [Google Scholar] [CrossRef]
  118. Baldoni, D.; Furustrand Tafin, U.; Aeppli, S.; Angevaare, E.; Oliva, A.; Haschke, M.; Zimmerli, W.; Trampuz, A. Activity of Dalbavancin, Alone and in Combination with Rifampicin, against Meticillin-Resistant Staphylococcus aureus in a Foreign-Body Infection Model. Int. J. Antimicrob. Agents 2013, 42, 220–225. [Google Scholar] [CrossRef]
  119. Muller-Serieys, C.; Saleh Mghir, A.; Massias, L.; Fantin, B. Bactericidal Activity of the Combination of Levofloxacin with Rifampin in Experimental Prosthetic Knee Infection in Rabbits Due to Methicillin-Susceptible Staphylococcus aureus. Antimicrob. Agents Chemother. 2009, 53, 2145–2148. [Google Scholar] [CrossRef] [Green Version]
  120. Landersdorfer, C.B.; Bulitta, J.B.; Kinzig, M.; Holzgrabe, U.; Sörgel, F. Penetration of Antibacterials into Bone: Pharmacokinetic, Pharmacodynamic and Bioanalytical Considerations. Clin. Pharm. 2009, 48, 89–124. [Google Scholar] [CrossRef]
  121. Sendi, P.; Zimmerli, W. Antimicrobial Treatment Concepts for Orthopaedic Device-Related Infection. Clin. Microbiol. Infect. 2012, 18, 1176–1184. [Google Scholar] [CrossRef] [Green Version]
  122. Lora-Tamayo, J.; Euba, G.; Cobo, J.; Horcajada, J.P.; Soriano, A.; Sandoval, E.; Pigrau, C.; Benito, N.; Falgueras, L.; Palomino, J.; et al. Short- versus Long-Duration Levofloxacin plus Rifampicin for Acute Staphylococcal Prosthetic Joint Infection Managed with Implant Retention: A Randomised Clinical Trial. Int. J. Antimicrob. Agents 2016, 48, 310–316. [Google Scholar] [CrossRef]
  123. Nguyen, S.; Robineau, O.; Titecat, M.; Blondiaux, N.; Valette, M.; Loiez, C.; Beltrand, E.; Migaud, H.; Senneville, E. Influence of Daily Dosage and Frequency of Administration of Rifampicin-Levofloxacin Therapy on Tolerance and Effectiveness in 154 Patients Treated for Prosthetic Joint Infections. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 1675–1682. [Google Scholar] [CrossRef]
  124. Wouthuyzen-Bakker, M.; Tornero, E.; Morata, L.; Nannan Panday, P.V.; Jutte, P.C.; Bori, G.; Kampinga, G.A.; Soriano, A. Moxifloxacin plus Rifampin as an Alternative for Levofloxacin plus Rifampin in the Treatment of a Prosthetic Joint Infection with Staphylococcus aureus. Int. J. Antimicrob. Agents 2018, 51, 38–42. [Google Scholar] [CrossRef] [PubMed]
  125. Fily, F.; Jolivet-Gougeon, A.; Polard, E.; Gicquel, T.; Dupont, M.; Verdier, M.C.; Arvieux, C. Moxifloxacin-Rifampicin Combination for the Treatment of Non-Staphylococcal Gram-Positive Orthopedic Implant-Related Infections. Med. Mal. Infect. 2019, 49, 540–544. [Google Scholar] [CrossRef] [PubMed]
  126. Meléndez-Carmona, M.Á.; Muñoz-Gallego, I.; Viedma, E.; Lora-Tamayo, J.; Chaves, F. Intraosteoblastic Activity of Levofloxacin and Rifampin Alone and in Combination against Clinical Isolates of Meticillin-Susceptible Staphylococcus aureus Causing Prosthetic Joint Infection. Int. J. Antimicrob. Agents 2019, 54, 356–360. [Google Scholar] [CrossRef] [PubMed]
  127. Rimmelé, T.; Boselli, E.; Breilh, D.; Djabarouti, S.; Bel, J.C.; Guyot, R.; Saux, M.C.; Allaouchiche, B. Diffusion of Levofloxacin into Bone and Synovial Tissues. J. Antimicrob. Chemother. 2004, 53, 533–535. [Google Scholar] [CrossRef] [Green Version]
  128. von Baum, H.; Böttcher, S.; Abel, R.; Gerner, H.J.; Sonntag, H.G. Tissue and Serum Concentrations of Levofloxacin in Orthopaedic Patients. Int. J. Antimicrob. Agents 2001, 18, 335–340. [Google Scholar] [CrossRef]
  129. Landersdorfer, C.B.; Kinzig, M.; Hennig, F.F.; Bulitta, J.B.; Holzgrabe, U.; Drusano, G.L.; Sörgel, F.; Gusinde, J. Penetration of Moxifloxacin into Bone Evaluated by Monte Carlo Simulation. Antimicrob. Agents Chemother. 2009, 53, 2074–2081. [Google Scholar] [CrossRef] [Green Version]
  130. Tsegka, K.G.; Voulgaris, G.L.; Kyriakidou, M.; Kapaskelis, A.; Falagas, M.E. Intravenous Fosfomycin for the Treatment of Patients with Bone and Joint Infections: A Review. Expert Rev. Anti Infect. Ther. 2021, 20, 33–43. [Google Scholar] [CrossRef]
  131. Schintler, M.V.; Traunmüller, F.; Metzler, J.; Kreuzwirt, G.; Spendel, S.; Mauric, O.; Popovic, M.; Scharnagl, E.; Joukhadar, C. High Fosfomycin Concentrations in Bone and Peripheral Soft Tissue in Diabetic Patients Presenting with Bacterial Foot Infection. J. Antimicrob. Chemother. 2009, 64, 574–578. [Google Scholar] [CrossRef] [Green Version]
  132. Pradier, M.; Robineau, O.; Boucher, A.; Titecat, M.; Blondiaux, N.; Valette, M.; Loïez, C.; Beltrand, E.; Nguyen, S.; Dézeque, H.; et al. Suppressive Antibiotic Therapy with Oral Tetracyclines for Prosthetic Joint Infections: A Retrospective Study of 78 Patients. Infection 2018, 46, 39–47. [Google Scholar] [CrossRef]
  133. Pradier, M.; Nguyen, S.; Robineau, O.; Titecat, M.; Blondiaux, N.; Valette, M.; Loïez, C.; Beltrand, E.; Dézeque, H.; Migaud, H.; et al. Suppressive Antibiotic Therapy with Oral Doxycycline for Staphylococcus aureus Prosthetic Joint Infection: A Retrospective Study of 39 Patients. Int. J. Antimicrob. Agents 2017, 50, 447–452. [Google Scholar] [CrossRef]
  134. Escudero-Sanchez, R.; Senneville, E.; Digumber, M.; Soriano, A.; Del Toro, M.D.; Bahamonde, A.; Del Pozo, J.L.; Guio, L.; Murillo, O.; Rico, A.; et al. Suppressive Antibiotic Therapy in Prosthetic Joint Infections: A Multicentre Cohort Study. Clin. Microbiol. Infect. 2020, 26, 499–505. [Google Scholar] [CrossRef] [PubMed]
  135. Leijtens, B.; Weerwag, L.; Schreurs, B.W.; Kullberg, B.-J.; Rijnen, W. Clinical Outcome of Antibiotic Suppressive Therapy in Patients with a Prosthetic Joint Infection after Hip Replacement. J. Bone Jt. Infect. 2019, 4, 268–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Wouthuyzen-Bakker, M.; Nijman, J.M.; Kampinga, G.A.; van Assen, S.; Jutte, P.C. Efficacy of Antibiotic Suppressive Therapy in Patients with a Prosthetic Joint Infection. J. Bone Jt. Infect. 2017, 2, 77–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Deconinck, L.; Dinh, A.; Nich, C.; Tritz, T.; Matt, M.; Senard, O.; Bessis, S.; Bauer, T.; Rottman, M.; Salomon, J.; et al. Efficacy of Cotrimoxazole (Sulfamethoxazole-Trimethoprim) as a Salvage Therapy for the Treatment of Bone and Joint Infections (BJIs). PLoS ONE 2019, 14, e0224106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Courjon, J.; Demonchy, E.; Cua, E.; Bernard, E.; Roger, P.-M. Efficacy and Safety of Clindamycin-Based Treatment for Bone and Joint Infections: A Cohort Study. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 2513–2518. [Google Scholar] [CrossRef] [PubMed]
  139. Zhuchenko, G.; Schmidt-Malan, S.; Patel, R. Planktonic and Biofilm Activity of Eravacycline against Staphylococci Isolated from Periprosthetic Joint Infections. Antimicrob. Agents Chemother. 2020, 64, e01304-20. [Google Scholar] [CrossRef]
  140. Dunne, M.W.; Puttagunta, S.; Sprenger, C.R.; Rubino, C.; Van Wart, S.; Baldassarre, J. Extended-Duration Dosing and Distribution of Dalbavancin into Bone and Articular Tissue. Antimicrob. Agents Chemother. 2015, 59, 1849–1855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Cojutti, P.G.; Rinaldi, M.; Zamparini, E.; Rossi, N.; Tedeschi, S.; Conti, M.; Pea, F.; Viale, P. Population Pharmacokinetics of Dalbavancin and Dosing Consideration for Optimal Treatment of Adult Patients with Staphylococcal Osteoarticular Infections. Antimicrob. Agents Chemother. 2021, 65, e02260-20. [Google Scholar] [CrossRef] [PubMed]
  142. Cojutti, P.G.; Rinaldi, M.; Gatti, M.; Tedeschi, S.; Viale, P.; Pea, F. Usefulness of Therapeutic Drug Monitoring in Estimating the Duration of Dalbavancin Optimal Target Attainment in Staphylococcal Osteoarticular Infections: A Proof-of-Concept. Int. J. Antimicrob. Agents 2021, 58, 106445. [Google Scholar] [CrossRef] [PubMed]
  143. Cojutti, P.G.; Merelli, M.; Bassetti, M.; Pea, F. Proactive Therapeutic Drug Monitoring (TDM) May Be Helpful in Managing Long-Term Treatment with Linezolid Safely: Findings from a Monocentric, Prospective, Open-Label, Interventional Study. J. Antimicrob. Chemother. 2019, 74, 3588–3595. [Google Scholar] [CrossRef] [PubMed]
  144. Pea, F.; Viale, P.; Cojutti, P.; Del Pin, B.; Zamparini, E.; Furlanut, M. Therapeutic Drug Monitoring May Improve Safety Outcomes of Long-Term Treatment with Linezolid in Adult Patients. J. Antimicrob. Chemother. 2012, 67, 2034–2042. [Google Scholar] [CrossRef] [PubMed]
  145. Pea, F.; Cojutti, P.; Pagotto, A.; Cristini, F.; Furlanut, M.; Viale, P. Successful Long-Term Treatment of Cerebral Nocardiosis with Unexpectedly Low Doses of Linezolid in an Immunocompromised Patient Receiving Complex Polytherapy. Antimicrob. Agents Chemother. 2012, 56, 3438–3440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Gatti, M.; Fusaroli, M.; Raschi, E.; Moretti, U.; Poluzzi, E.; De Ponti, F. Serious Adverse Events with Tedizolid and Linezolid: Pharmacovigilance Insights through the FDA Adverse Event Reporting System. Expert Opin. Drug Saf. 2021, 20, 1421–1431. [Google Scholar] [CrossRef] [PubMed]
  147. Esposito, S.; Leone, S.; Noviello, S.; Ianniello, F.; Fiore, M.; Russo, M.; Foti, G.; Carpentieri, M.S.; Cellesi, C.; Zanelli, G.; et al. Outpatient Parenteral Antibiotic Therapy for Bone and Joint Infections: An Italian Multicenter Study. J. Chemother. 2007, 19, 417–422. [Google Scholar] [CrossRef]
  148. Allen, J.; Adams, K.; Thompson, F.; Cullen, L.; Barlow, G. Long-Term, Once-Weekly Outpatient Teicoplanin Use for Suppression of Chronic Prosthetic Joint Infection. Int. J. Antimicrob. Agents 2013, 41, 200–201. [Google Scholar] [CrossRef] [PubMed]
  149. Gatti, M.; Bianchin, M.; Raschi, E.; De Ponti, F. Assessing the Association between Fluoroquinolones and Emerging Adverse Drug Reactions Raised by Regulatory Agencies: An Umbrella Review. Eur. J. Intern. Med. 2020, 75, 60–70. [Google Scholar] [CrossRef] [PubMed]
  150. Schwarz, E.M.; McLaren, A.C.; Sculco, T.P.; Brause, B.; Bostrom, M.; Kates, S.L.; Parvizi, J.; Alt, V.; Arnold, W.V.; Carli, A.; et al. Adjuvant Antibiotic-loaded Bone Cement: Concerns with Current Use and Research to Make It Work. J Orthop Res 2021, 39, 227–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Abosala, A.; Ali, M. The Use of Calcium Sulphate Beads in Periprosthetic Joint Infection, a Systematic Review. J. Bone Jt. Infect. 2020, 5, 43–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. De Meo, D.; Ceccarelli, G.; Iaiani, G.; Lo Torto, F.; Ribuffo, D.; Persiani, P.; Villani, C. Clinical Application of Antibacterial Hydrogel and Coating in Orthopaedic and Traumatology Surgery. Gels 2021, 7, 126. [Google Scholar] [CrossRef] [PubMed]
  153. De Meo, D.; Cannari, F.M.; Petriello, L.; Persiani, P.; Villani, C. Gentamicin-Coated Tibia Nail in Fractures and Nonunion to Reduce Fracture-Related Infections: A Systematic Review. Molecules 2020, 25, 5471. [Google Scholar] [CrossRef] [PubMed]
  154. Bhimani, S.; Carli, A.; Sethuraman, A.; Yang, X.; Ross, F.P.; Bostrom, M.P. The Novel Antibiotic Dalbavancin Is Heat Stable, Elutes from PMMA Cement Spacers and Has Superior Antimicrobial Activity than Vancomycin in an in-Vitro Killing Assay of Staphylococcus aureus. 2017. Available online: http://www.ors.org/Transactions/63/1218.pdf (accessed on 10 January 2022).
  155. De Meo, D.; Calogero, V.; Are, L.; Cavallo, A.U.; Persiani, P.; Villani, C. Antibiotic-Loaded Hydrogel Coating to Reduce Early Postsurgical Infections in Aseptic Hip Revision Surgery: A Retrospective, Matched Case-Control Study. Microorganisms 2020, 8, 571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Murad, M.H.; Asi, N.; Alsawas, M.; Alahdab, F. New Evidence Pyramid. Evid. Based Med. 2016, 21, 125–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The major determinants involved in the choice of the best antibiotic strategy for the management of orthopaedic implant-associated infections.
Figure 1. The major determinants involved in the choice of the best antibiotic strategy for the management of orthopaedic implant-associated infections.
Antibiotics 11 00406 g001
Figure 2. The role of biofilm formation and the major determinants involved in the choice of antibiofilm agents. CoNS: coagulase-negative Staphylococci; MBBC: minimal biofilm bactericidal concentration; MBEC: minimal biofilm eradication concentration; MBIC: minimal biofilm inhibitory concentration; PK/PD: pharmacokinetic/pharmacodynamic SCV: small colony variants.
Figure 2. The role of biofilm formation and the major determinants involved in the choice of antibiofilm agents. CoNS: coagulase-negative Staphylococci; MBBC: minimal biofilm bactericidal concentration; MBEC: minimal biofilm eradication concentration; MBIC: minimal biofilm inhibitory concentration; PK/PD: pharmacokinetic/pharmacodynamic SCV: small colony variants.
Antibiotics 11 00406 g002
Figure 3. The features of selected anti-staphylococcal agents in relationship with major determinants required for the management of orthopaedic implant-associated infections. Green box: optimal activity/efficacy; yellow box: some concerns in activity/efficacy; red box: limited or absent activity/efficacy.
Figure 3. The features of selected anti-staphylococcal agents in relationship with major determinants required for the management of orthopaedic implant-associated infections. Green box: optimal activity/efficacy; yellow box: some concerns in activity/efficacy; red box: limited or absent activity/efficacy.
Antibiotics 11 00406 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gatti, M.; Barnini, S.; Guarracino, F.; Parisio, E.M.; Spinicci, M.; Viaggi, B.; D’Arienzo, S.; Forni, S.; Galano, A.; Gemmi, F. Orthopaedic Implant-Associated Staphylococcal Infections: A Critical Reappraisal of Unmet Clinical Needs Associated with the Implementation of the Best Antibiotic Choice. Antibiotics 2022, 11, 406. https://doi.org/10.3390/antibiotics11030406

AMA Style

Gatti M, Barnini S, Guarracino F, Parisio EM, Spinicci M, Viaggi B, D’Arienzo S, Forni S, Galano A, Gemmi F. Orthopaedic Implant-Associated Staphylococcal Infections: A Critical Reappraisal of Unmet Clinical Needs Associated with the Implementation of the Best Antibiotic Choice. Antibiotics. 2022; 11(3):406. https://doi.org/10.3390/antibiotics11030406

Chicago/Turabian Style

Gatti, Milo, Simona Barnini, Fabio Guarracino, Eva Maria Parisio, Michele Spinicci, Bruno Viaggi, Sara D’Arienzo, Silvia Forni, Angelo Galano, and Fabrizio Gemmi. 2022. "Orthopaedic Implant-Associated Staphylococcal Infections: A Critical Reappraisal of Unmet Clinical Needs Associated with the Implementation of the Best Antibiotic Choice" Antibiotics 11, no. 3: 406. https://doi.org/10.3390/antibiotics11030406

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop