Next Article in Journal
Immunomodulatory, Anticancer, and Antimicrobial Effects of Rice Bran Grown in Iraq: An In Vitro and In Vivo Study
Next Article in Special Issue
Changes in Pharmacokinetics and Pharmacodynamics of Losartan in Experimental Diseased Rats Treated with Curcuma longa and Lepidium sativum
Previous Article in Journal
Antiviral Effect of Stenocline ericoides DC. and Stenocline inuloides DC., Two Flavonoid-Rich Endemic Plants from Madagascar, against Dengue and Zika Viruses
Previous Article in Special Issue
Aspartames Alter Pharmacokinetics Parameters of Erlotinib and Gefitinib and Elevate Liver Enzymes in Wistar Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Assessing Clinical Potential of Old Antibiotics against Severe Infections by Multi-Drug-Resistant Gram-Negative Bacteria Using In Silico Modelling

1
Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
2
Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 CN Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(12), 1501; https://doi.org/10.3390/ph15121501
Submission received: 15 October 2022 / Revised: 10 November 2022 / Accepted: 16 November 2022 / Published: 30 November 2022

Abstract

:
In the light of increasing antimicrobial resistance among gram-negative bacteria and the lack of new more potent antimicrobial agents, new strategies have been explored. Old antibiotics, such as colistin, temocillin, fosfomycin, mecillinam, nitrofurantoin, minocycline, and chloramphenicol, have attracted the attention since they often exhibit in vitro activity against multi-drug-resistant (MDR) gram-negative bacteria, such as Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii. The current review provides a summary of the in vitro activity, pharmacokinetics and PK/PD characteristics of old antibiotics. In silico modelling was then performed using Monte Carlo simulation in order to combine all preclinical data with human pharmacokinetics and determine the probability of target (1-log kill in thigh/lung infection animal models) attainment (PTA) of different dosing regimens. The potential of clinical efficacy of a drug against severe infections by MDR gram-negative bacteria was considered when PTA was >95% at the epidemiological cutoff values of corresponding species. In vitro potent activity against MDR gram-negative pathogens has been shown for colistin, polymyxin B, temocillin (against E. coli and K. pneumoniae), fosfomycin (against E. coli), mecillinam (against E. coli), minocycline (against E. coli, K. pneumoniae, A. baumannii), and chloramphenicol (against E. coli) with ECOFF or MIC90 ≤ 16 mg/L. When preclinical PK/PD targets were combined with human pharmacokinetics, Monte Carlo analysis showed that among the old antibiotics analyzed, there is clinical potential for polymyxin B against E. coli, K. pneumoniae, and A. baumannii; for temocillin against K. pneumoniae and E. coli; for fosfomycin against E. coli and K. pneumoniae; and for mecillinam against E. coli. Clinical studies are needed to verify the potential of those antibiotics to effectively treat infections by multi-drug resistant gram-negative bacteria.

1. Introduction

The ever-increasing antimicrobial resistance worldwide poses an urgent threat to our antimicrobial arsenal. In particular, gram-negative bacteria, such as Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii, rapidly develop resistance to many currently licensed antimicrobial drugs, including recently introduced drugs. According to the ECDC 2022 report, only 46% of E. coli, 62% of K. pneumoniae, 70% of P. aeruginosa, and 34% of A. baumannii were fully susceptible to all antimicrobial drugs [1]. Resistance rates to at least two antimicrobial groups were 10.2% for E. coli, 15.7% for K. pneumoniae, 7.6% for P. aeruginosa, and 57.3% for A. baumannii. The rates of multi-drug resistance (MDR) phenotypes are increasing every year, limiting therapeutic options. Of particular concern are extended-spectrum β-lactamase-producing Enterobacterales, carbapenem-resistant A. baumannii and Enterobacterales, and MDR P. aeruginosa, with the pipeline of new antimicrobial agents being very limited due to the time-consuming process and the exorbitant costs for the development of new and hopefully potent drugs [2]. As some old antibiotics introduced in 1950s show in vitro activity against those MDR bacteria, there has been an increased interest in those compounds as an alternative approach to treating MDR infections [3,4,5].
Among the old antimicrobial agents, some were neglected because new drugs have been introduced with improved activity and safety profiles and convenient administration routes. Among them, the most interesting compounds from a clinical point of view were considered to be colistin, temocillin, mecillinam, nitrofurantoin, fosfomycin, minocycline, and chloramphenicol [6]. Those drugs never underwent the processes that new drugs now undergo for drug efficacy assessment and regulatory approval. These agents are both cheap with broad spectrum activity, and some of them have been used successfully to treat non-severe infections, such as urinary tract infections, by MDR. One of the major advantages of old antibiotics is the fact that they are not currently widely used, and therefore, resistance levels are expected to be low [7].
However, data regarding the probability of therapeutic success and the appropriate dose for severe infections, such as bloodstream infections and pneumonia are sparse. It is imperative to assess these drugs based on more up-to-date pharmacokinetic/pharmacodynamic (PK/PD) studies and explore any potential for use against MDR infections. Old drugs were only recently assessed based on current guidelines on the use of pharmacokinetics (PKs) and pharmacodynamics (PDs) for the development of antimicrobial medicinal products. In silico modelling is an important tool for assessing whether licensed or alternative dosing regimens of those drugs can attain preclinical PK/PD targets [8]. We therefore reviewed in vitro susceptibility, PK, and PK/PD data of old drugs and performed in silico modelling in order to estimate the probability of target attainment (PTA) ousing previously published PK/PD targets.

2. Methods

In vitro susceptibility, PK and PK/PD data of colistin, temocillin, mecillinam, nitrofurantoin, fosfomycin, minocycline, and chloramphenicol against E. coli, K. pneumoniae, for P. aeruginosa, and A. baumannii—particularly MDR pathogens—were reviewed. Monte Carlo simulation was then performed in order to bridge in vitro susceptibility data, preclinical PK/PD targets, and human PKs and determine the PTA for each drug and gram-negative MDR species. Monte Carlo simulation analysis is a well-established approach to simulating exposures in a large number of patients and associated variation based on mean and standard deviation (SD) PK parameters derived from a small cohort of patients in clinical PK studies. For this reason, we used the KinFun 1.02 software (Maastricht, Netherlands) with the following input parameters: N, number of compartments; fu, unbound fraction; V1, volume central compartment; k10, (CL/V1) elimination rate constant; and k12 (Q/V1) and k21 (Q/V2), distribution rate constants, where Q is the intercompartmental clearance, V2 volume peripheral compartment, CL clearance. Rate constants were extracted from the literature; if there were no published reports, they were calculated based on reported CL and volume of distribution (Vd) values. SDs were used to add variability in CL and Vd. A log normal distribution was assumed for both CL and Vd in order to calculate individual rate constants for 5000 patients. Apart from that, route of administration, maintenance dose, number of doses, dosing time intervals, and infusion duration were taken into account in order to simulate PK profiles. Simulated CL, VD, and their corresponding SDs were compared to published values, allowing <3% deviation for all parameters. For each simulated patient, the PK/PD indices fAUC/MIC (area under the unbound plasma concentration time curve over the minimum inhibitory concentrations MIC), fCmax/MIC (peak of unbound plasma concentration over MIC), and %fT > MIC (% of dosing interval that unbound plasma concentrations remains above the MIC) were calculated for different MICs, taking into account the unbound fraction of drugs, as this fraction is considered pharmacodynamically important. Furthermore, the number of simulated patients attaining preclinical PK/PD targets corresponding to 1-log kill were plotted against MICs together with the MIC distribution from the European Committee on Antimicrobial Susceptibility Testing (EUCAST) website (www.eucast.org accessed on 23 October 2022) and, when not available, from previously published papers. The 1-log kill effect was chosen as it is the most relevant endpoint for severe infections, such as bloodstream infections and pneumonia [9]. Therefore, only preclinical data from thigh and lung infection models were utilized for determining PK/PD targets and when not available, data from in vitro dynamic models were used. The PK/PD target of 3-log kill was used from in vitro models as this seems to correlate with 1-log kill in animals [10].
The process consisted of 1. simulation of a virtual patient population infected with pathogens with increasing MICs, 2. calculation of PK/PD indices for each patient and each pathogen, 3. estimation of % of patients attaining the PK/PD target of 1-log kill effect for each MIC, 4. comparison of the MICs with a PTA > 95% with the epidemiological cutoff value (ECOFF), or MIC90 when ECOFF was not available for each species. In addition, the cumulative fraction of response (CFR) [11] was estimated for each antibiotic dosing regimen against MIC distributions of Enterobacterales, P. aeruginosa, and A. baumannii, as presented on the EUCAST website (www.eucast.org accessed on 14 October 2022) or in previously published papers. CFR was defined as the cumulative PTA of an expected population for a specific dosing regimen and a specific population of microorganisms and was calculated based on the equation CFR = ΣPTAixFi [11]: where the subscript i indicates the MIC value, ranked from highest to lowest MIC of the tested population of microorganisms; PTAi is the probability of target attainment of each MIC; and Fi is the fraction of microorganism population at each MIC category.
A drug was considered promising against a specific pathogen when the PTAs were >95% for the wild-type (WT) population, i.e., at the ECOFF (or the lowest concentration of antibiotic at which 90% of the isolates were inhibited (MIC90) if ECOFF has not been determined).

3. Colistin

Colistin (polymyxin E) is a member of the polymyxin group of antimicrobial compounds, which consist of basic polypeptide antibiotics with a side chain terminated by fatty acids. Colistin and polymyxin B were introduced to the pipeline of antibiotics for clinical use with similar antibacterial spectra. Colistin acts by displacing calcium and magnesium from the negatively charged lipopolysaccharide in the cell membrane of bacteria, leading to increased permeability of the cell envelope, loss of integrity of the membrane, leakage of cell contents, and finally, cell death. Acquired resistance to colistin is mainly due to lipopolysaccharide modifications [12].
Colistin is highly active against Enterobacterales, except Proteae and Serratia spp. It is also active against Pseudomonas aeruginosa and Acinetobacter spp. Based on EUCAST MIC distribution, colistin is highly potent against various gram-negative bacteria, with ECOFFs of 2, 2, 4, and 2 mg/L for E. coli, K. pneumoniae, P. aeruginosa, and A. baumannii, respectively (www.eucast.org accessed on 23 October 2022) (Table 1).
Colistin is administered to patients intravenously (IV) as the inactive prodrug colistin methanesulphonate sodium (CMS), which is converted to its active form in vivo. It is used for the treatment of infections caused by isolates that are resistant to less toxic antimicrobials. Moreover, patients with chronic bronchopulmonary colonization by P. aeruginosa are treated with colistin in inhaled form [12]. The most often used clinical IV dosing regimens are 9MIU q24h, 4.5MIU q12h, and 3MIU q8h, with tAUC0–24 50.18–72.93 mg∙h/L and tCmax 2.98–5.83 mg/L [39] (Table 2). The protein binding of colistin in plasma of 66 critically ill patients (and healthy humans) was found to be 40% (unbound fraction 0.5) [40]. Large fluctuations have been found in t1/2 ranging between 2–14.4 h [41,42].
The PK/PD index linked to antibacterial effect is the ratio fAUC/MIC [39]. fAUC/MIC values of 20.37 ± 4.13 and 14.83 ± 2.35 were previously found to correspond to 1-log kill of P. aeruginosa in thigh and lung infection animal models, respectively [58] (Table 3). Lower PK/PD indices were found in another study in thigh infection animal models (6.6–10.9 and 3.5–13.9 fAUC/MIC was associated with 1-log kill of P. aeruginosa and A. baumannii, respectively), probably because the unbound fraction for colistin was estimated to be lower (concentration-independent fu 0.084) compared to the previous studies (concentration-dependent fu 0.1–0.5). Thus, the PK/PD target for A. baumannii was underestimated, and for that reason, we used the strictest target found for the study above (fAUC/MIC 13.9) [40]. Regarding K. pneumoniae, in an In vitro PK/PD model, an fAUC/MIC 24 corresponded to a bactericidal effect of 3-log kill [39].
Monte Carlo simulation of 5000 patients was performed with a mean ± SD fAUC0–24 of 29.17 ± 15.44 for 9MU q24h (45 min IV infusion), 24.28 ± 4.8 for 4.5MU q12h (45min IV infusion), and 20.07 ± 5.37 for 3MU q8h (45 min IV infusion) [43] and using the mean PK/PD indices 20.37 for P. aeruginosa, 13.9 for A. baumannii, and 24 for K. pneumoniae. The PTAs were low at ECOFF of 4, 2 and 2 mg/L for P. aeruginosa, A. baumannii and K. pneumoniae for all dosing regimens tested, respectively (Figure 1). Considering the same PK/PD target of fAUC/MIC 24 as in K. pneumoniae, PTAs were <10% at ECOFF 2 mg/L for E. coli isolates for all dosing regimens tested. Hence, colistin seems not be a promising agent as monotherapy at the abovementioned doses against the MDR gramm(-)bacteria. This was confirmed by an open-label randomized controlled trial study; clinical failure rates ranged between 62–83% in patients treated with colistin monotherapy against infections caused by Enterobacterales, P. aeruginosa, and A. baumanii [66].

4. Polymyxin B

Polymyxin B is a cationic polypeptide antibiotic with very similar chemical structure to colistin (Polymyxin E), and it is mainly used for the treatment of infections caused by gram-negative bacteria—in particular, MDR K. pneumoniae, A. baumannii, and P. aeruginosa [67]. The mechanism of action involves the ability to bind with and disorganize the outer membrane of gram-negative bacteria, disrupting the osmotic equilibrium [68]. Resistance mechanisms include intrinsic and adaptive resistance via mutations of LPS, whereas horizontally acquired resistance mechanisms have not been reported [68]. Based on previous studies, polymyxin’s B MIC90 was 0.25 mg/L for E. coli, 0.5 mg/L for K. pneumoniae, 2 mg/L for P. aeruginosa, and 0.25–0.5 mg/L for A. baumannii isolates (Table 1) [18,22,23]. Higher polymyxin B MIC90 values were observed for carbapenem-resistant (CR) K. pneumoniae (MIC90 2–32 mg/L), although no information on previous treatment and resistance mechanisms was provided [20].
Polymyxins are very large lipopeptide molecules and are poorly absorbed via oral administration [69]. They are commonly given IV for the treatment of life-threatening systemic infections or by nebulization for the treatment of respiratory tract infections [70]. Pharmaceutical products contain active polymyxin B as sulphate salt [67]. Although polymyxin B is a potent antimicrobial compound, its clinical utility is widely limited by its potential for nephrotoxicity and neurotoxicity [28,71]. In general, dosage is based on total body weight, but PK data are not explored in all group of patients [72]. Due to significant knowledge gaps, the clinical dosing strategies of polymyxin B are not fully optimized, and for that reason, a thorough understanding of PKs is crucial to maximizing efficacy and minimizing toxicity [73].
The current understanding of polymyxin B PKs consists of four studies comprised of total 60 patients in total [74]. Clinically administered doses usually result with maximum serum concentration at steady state ranging from 2–14 μg/mL with a half-life of 9–11.5 h [75]. PKs of polymyxin B in different group of patients at clinical dosing regimens of 40–50 mg q12h, 119 ± 36.3 mg/day and 0.45–3.38 mg/kg/day resulted in mean ± SD AUC0–24 values of 74.6 ± 17.81, 52.3 ± 14.8, and 66.9 ± 21.6 mg·h/L, respectively (Table 2). The unbound fraction of polymyxin B in plasma of 24 critically ill patients was found to be 42% [46]. Half-life of the drug (t1/2) ranged between 10.1 and 11.9 h.
According to the findings of previous PK/PD studies, the PK/PD index that best describes the bactericidal activity of polymyxin B is AUC0–24/MIC [44]. fAUC/MIC values of 39.7 ± 14.4 and 50.6 ± 3.8 were associated with 1-log kill of K. pneumoniae and E. coli, respectively, in a thigh infection animal model (Table 3) [60]. Monte Carlo simulation analysis of 5000 patients was performed with mean ± SD fAUC0–24 31.33 ± 7.48, 21.96 ± 6.21, and 28.09 ± 9.07 for dosing regimens of 40–50 mg q12h, 119 ± 36.3 mg/day and 0.45–3.38 mg/kg/day, respectively. The PTA was >95% with the dosing regimen of 40–50 mg q12h in renal transplant patients for most E. coli (MIC90 0.25 mg/L) and K. pneumoniae (MIC90 0.5 mg/L) isolates (Figure 2). Considering the same PK/PD target for the other gram-negatives, the PTAs are expected to be high (>95%) for most A. baumannii isolates, as their MIC90 is ≤0.5 mg/L, but not for P. aeruginosa (MIC90 2 mg/L) or CRE K. pneumoniae (MIC90 2-32 mg/L) for all three dosing regimens tested. Indeed, IV polymyxin B therapy was inferior to other drugs in the treatment of P. aeruginosa bacteremia and K. pneumoniae [76,77], whereas the use of polymyxin B was effective against A. baumannii infections [78]. For that reason, polymyxin B seems to be quite promising against E. coli, K. pneumoniae, and A. baumannii but not against P. aeruginosa infections. Studies assessing clinical efficacy of polymyxin B against multi-drug-resistant gram-negative bacteria found lower mortality for A. baumannii than P. aeruginosa infections, although in most cases, polymyxin B was given in combination with other drugs [78,79].

5. Temocillin

Temocillin, a 6-a-methoxy derivative of ticarcillin, is a penicillin with restricted spectrum against Enterobacterales, while non-fermenters, anaerobes and gram-positive bacteria are not within its spectrum [80]. The addition of α-methoxy moiety on ticarcillin prevents hydrolysis against a wide variety of β-lactamases, including extended spectrum β-lactamases (ESBLs) [81], ampicilinases C (AmpCs) [3] and some carbapenemases [82], but not metallo-beta-lactamases (MBL) and oxacillininases (OXAs) [3,29]. The mechanism of action relies on the prevention by α-methoxy group of entry of a water molecule into the β-lactamase active site, which leads to hydrolysis by preventing activation of the serine and other chemical events [3]. On the other hand, the intrinsic resistance of P. aeruginosa isolates to temocillin is mainly due to active efflux by the constitutively expressed MexAB-OprM efflux transporters [83]. In addition, there are in vitro studies that demonstrate the efficacy of active drugs against ESBL-producing strains, with susceptibility rates of up to 80% and 90% using breakpoints of 8 and 16 mg/L, respectively [26,81,84,85]. In addition, an observational study supports the clinical use of temocillin as a potential alternative to carbapenems against infections caused by ESBL-/AmpC-producing Enterobacterales [86]. Based on EUCAST distribution, temocillin is highly potent against various gram-negative bacteria, with ECOFF being 16 and 8 mg/L for E. coli and K. pneumoniae, respectively (www.eucast.org). The MIC90 of temocillin was ≥256 mg/L against P. aeruginosa and A. baumannii (Table 1) [83,87].
Temocillin can only be administered parenterally (IV, intramuscularly, and subcutaneously), and it is used for empirical treatment of pyelonephritis and complicated UTI [57]. It is bactericidal, has a prolonged elimination half-life of approximately 5 h, and has a high percentage of protein binding (~80%). Clearance of temocillin is mainly renal, and urinary recovery ranges from 72–82% after 24 h. Moreover, it has high penetration into bile and peritoneal fluid but poor penetration into cerebrospinal fluid. PKs of temocillin were simulated in different dosing regimens and different groups of patients, as shown in Table 2. Briefly, clinical dosing regimens of 2 g q12h (4 g/24 h) and 2 g q8h (6 g/24 h) result in AUC0–24 values of 1856 ± 282 and 1764 mg·h/L, and Cmax values of 147 ± 12 and 170 mg/L, respectively. The percentage of free drug was 23.7 ± 6.15% [47]. The half-life of the drug was 4.3 ± 0.3 h.
The PK/PD index correlating with temocillin efficacy seems to be fT > MIC. %fT > MIC values of 81.5 ± 14.4 and 79 ± 6.4 were associated with 1-log kill of E. coli and K. pneumoniae in thigh and 35 ± 18.3 and 47.3 ± 21.4 for lung infection animal models, respectively [61] (Table 3). Monte Carlo simulation of 5000 patients with mean ± SD Cl 2.44 ± 0.39 L/h and VD 14.3 ± 0.87 L for 2g q12h, and with mean ± SD Cl 3.69 ± 0.45 L/h, V1 14 ± 2.51 and V2 21.7 ± 4.52 for 2g q8h was performed using KinFun. For the Monte Carlo analysis, an unbound fraction of 24% has been used for both dosing regimens. For E. coli, the PTAs of the 1-log kill PK/PD target in the thigh infection model were low (<50%) at the ECOFF of 16 mg/L with the 2g q12h and 2g q8h dosing regimens, whereas the PTAs of 1-log kill PK/PD target in lung infection model were >90% at the ECOFF of 16 mg/L with a 2g q8h dosing regimen (Figure 3). For K. pneumoniae, the PTA of the PK/PD target in thigh infection model was <85% at the ECOFF of 8 mg/L for both dosing regimens, whereas the PTA of the PK/PD target in the lung infection model was 100% at the ECOFF of 8 mg/L with both dosing regimens. If the PK/PD targets were the same for P. aeruginosa and A. baumannii, PTAs are expected to be low for WT isolates with 2g q12h and 2g q8h dosing regimens due to high MIC90 (≥256 mg/L) for both species [29]. Providing that the same tissue penetration occurs in humans, the 2g q12h and 2g q8h dosing regimens are promising for pneumonia by E. coli and K. pneumoniae.
In a large, multicenter, retrospective, open, noncomparative study, clinical/microbiological efficacies of temocillin against infections by Enterobacterales (55% E. coli, 14% K. pneumoniae) were 83%/82% for BSI and 75%/67% for hospital-acquired pneumonia (HAP), being slightly lower than the 90%/87% in urinary tract infection (UTI) [86]. In a single-center, retrospective study, clinical failure was higher in non-UTI than UTI (26.7% vs. 4.9%) and in patients with septic shock compared to patient with sepsis (25% vs. 6.2%) by Enterobacterales (mainly E. coli), with no difference observed between the 2g q12h and 2g q8h dosing regimens, although q8h regimen was more frequently given in seriously ill patients [88]. Interestingly, significantly fewer failures were observed for K. pneumoniae infections. Thus, temocillin may have a role in treating E. coli and K. pneumoniae infections.

6. Fosfomycin

Fosfomycin, first isolated from Streptomyces spp. in 1969, is an organic phosphonate agent. It is the smallest molecule among all antimicrobials (138 Da), and cross-resistance with other classes is rare. It is rapidly bactericidal, inhibiting cell wall synthesis via irreversible inhibition of the enol-pyruvyl transferase. Penetration into the bacterial cell occurs through two different active transport systems, the inducible and predominant hexose monophosphate route which operates in the presence of glucose-6-phosphate inducer (GlpT) and the constitutive L-α-glycerophosphate system (UhpT) [89]. Concerning resistance mechanisms to fosfomycin, there are two intrinsic and one acquired mechanisms. For intrinsic mechanisms, inactivation of fosfomycin occurs via cleavage of the molecule by bacterial Fos enzymes. The majority of K. pneumoniae, K. aerogenes, P. aeruginosa, and Enterobacter spp. have FosA enzymes. Moreover, fosfomycin inhibits MurA, which initiates peptidoglycan biosynthesis of the bacterial cell wall. Resistance to fosfomycin in several bacteria is common mainly through MurA mutations, to which fosfomycin must bind to exhibit its antibacterial effect. Acquired resistance to fosfomycin occurs through modifications of membrane transporters GlpT and UhpT preventing active drug entering the bacterial cell, resulting in reduced uptake of fosfomycin by the pathogen [89]. Although resistance rates in clinical isolates are still relatively low, the emergence of resistance occurs rapidly in vitro. Resistant mutants arise in vitro at a frequency of 10−4 to 10−5. In ESBL-producing E. coli, in vivo resistance is increasingly recognized [90].
Fosfomycin has a broad spectrum of activity including both gram-negative and gram-positive bacteria and recently gained considerable attention due to its effectiveness against multi-drug-resistant pathogens [91], including ESBL and carbapenemase-producing isolates. Based on EUCAST distribution, fosfomycin is highly potent against Enterobacterales but seems less effective against P. aeruginosa and A. baumannii isolates. The ECOFF for E. coli, K. pneumoniae, and P. aeruginosa are 4, 128, and 256 mg/L, respectively (www.eucast.org, accessed on 23 October 2022). High MIC90 (>256 mg/L) has been reported for carbapenemase-producing Enterobacterales (Table 1). Fosfomycin resistance among carbapenemase-producing Enterobacterales is an emerging problem and is due to the plasmid-mediated fosfomycin resistance gene fosA3 and mutation in the transporter glpT [92].
Fosfomycin is hydrophilic with negligible protein binding and is eliminated exclusively by glomerular filtration, so its clearance depends mainly on the patient’s renal function. Regarding the volume of distribution, it is approximately 0.3 L/kg, but in critically ill patients suffering from bacterial infections, it is increased [93]. Moreover, fosfomycin is well tolerated and exhibits extensive penetration into many tissues with minor side effects reported [94]. PKs of fosfomycin were simulated in non-critically-ill patients based on the most recent study, as is shown in Table 2. The clinical dosing regimen of 4 g q6h resulted in a mean ± SD fAUC0–24 values of 5215 ± 1972.2 mg∙h/L the first 24 h [50] although lower exposures have been described in other studies [90]. Fosfomycin does not bind to plasma proteins [90]. The half-life of the drug ranged between 2.41–12.1 h depending on the route of administration [91].
Fosfomycin is hydrophilic with negligible protein binding and is eliminated exclusively by glomerular filtration, so its clearance depends mainly on the patient’s renal function. Regarding the volume of distribution, it is approximately 0.3 L/kg, but in critically ill patients suffering from bacterial infections, it is increased [93]. Moreover, fosfomycin is well tolerated and exhibits extensive penetration into many tissues with minor side effects reported [94]. PKs of fosfomycin were simulated in non-critically-ill patients based on the most recent study, as is shown in Table 2. The clinical dosing regimen of 4 g q6h resulted in a mean ± SD fAUC0–24 values of 5215 ± 1972.2 mg∙h/L the first 24 h [50] although lower exposures have been described in other studies [90]. Fosfomycin does not bind to plasma proteins [90]. The half-life of the drug ranged between 2.41–12.1 h depending on the route of administration [91].
The optimal PK/PD index characterizing fosfomycin activity is AUC/MIC, as found in dose fractionation studies in animals (R2 = 0.70 for AUC/MIC, R2 = 0.51 for Cmax/MIC, R2 = 0.44 for T > MIC) [62]. Monte Carlo simulation analysis of 5000 patients was performed for 4 g q8h (12 g/d), 6 g q8h (18 g/d), and 8 g q8h (24 g/d). The fAUC0–24 was calculated based on the fAUC0–24 of 5215 ± 1972.2 mg∙h/L for the 4g q6h (16 g/d) as described by Merino-Bohorquez et al. [50], assuming linear PKs and the same variation across different total daily doses, with mean ± SD fAUC0–24 3911 ± 1479 mg∙h/L for 4 g q8h (12 g/d) dosing regimen, 5867 ± 2219 mg∙h/L for 6 g q8h (18 g/d), and 7823 ± 2958 mg∙h/L for 8 g q8h (24 g/d). Mean ± SD AUC0–24/MIC values of 98.9 ± 78.4, 21.5, and 28.2 ± 17.82 corresponded to 1-log kill of E. coli, K. pneumoniae, and P. aeruginosa, respectively were previously found in thigh infection animal models [62] (Table 3). The PTAs were 100% at the ECOFF of 4 mg/L for E. coli for all simulated dosing regimens tested (Figure 4). This also can be confirmed by the high rate of CFR (>95%) in all dosing regimens. The PTAs were high (>95%) at the ECOFF of K. pneumoniae isolates only for dosing regimens with 18g/day, while for Klebsiella pnemoniae carbapenemase (KPC), New Delhi metallo-β-lactamase (NDM), MBL, OXA-48-producing isolates with MIC90 256 mg/L, and for WT P. aeruginosa (MIC ≤ 256 mg/L) isolates, none of the dosing regimens could be effective (PTA ≤ 74%). Fosfomycin has been rarely used as monotherapy to treat severe infections by MDR pathogens. In a multi-center, randomized, double-blind comparative study (ZEUS study), fosfomycin was given as monotherapy mainly against UTI, and clinical cure was observed in 25/27 infections caused by K. pneumoniae, 8/8 caused by P. aeruginosa, 120/133 caused by E. coli, and 2/2 caused by A. baumannii [95]. Thus, there is a clinical potential for fosfomycin against E. coli and K. pneumoniae, although the high MICs reported for carbapenemase-producing Enterobacterales may limit fosfomycin coverage.

7. Mecillinam

Mecillinam, or 6β-amidinopenicillanic acid, is an amidinopenicillin developed in 1972 and has been used extensively in Scandinavian countries for the treatment of acute lower UTI caused by Enterobacterales, and especially by E. coli, since the 1980s [96]. The antimicrobial is detected in high concentrations in urine, and its impact on the intestinal microbiota was found to be low [97,98]. The antimicrobial agent is administered orally as pivmecillinam, which is hydrolyzed to the active drug in vivo. The prodrug pivmecillinam is a unique β-lactam with high specificity against penicillin-binding protein 2 (PBP-2) in gram-negative cell walls, and extensive activity against Enterobacterales, that also resists hydrolysis by β-lactamases [5]. Biochemical and genetic studies revealed that mecillinam interacts with PBP-2, resulting in the production of round, osmotically stable bacterial cells [99]. Resistance development is associated with mutations in a large number of genes that affect many different cellular functions, including cell elongation and division, composition of lipopolysaccharide in combination with cya/crp, and cysteine biosynthesis [100]. Evaluating the in vitro efficacy of the drug, remarkable activity is retained against ESBL and AmpC β-lactamases. Moreover, recent data suggest that mecillinam is frequently active in vitro against NDM and imipenemase (IMP) metallo-β-lactamases and OXA-48 producers but not against KPC and Verona integron-encoded metallo-β-lactamase (VIM) [3,100]. Based on EUCAST MIC distribution, mecillinam seems to be potent against Enterobacterales isolates. The MIC90 values were 2 mg/L and 128 mg/L for E. coli and K. pneumoniae, respectively, with a tentative ECOFF of 0.5 mg/L for E. coli (Table 1). Mecillinam is inactive against P. aeruginosa and A. baumannii (MIC > 128 mg/L) [101,102].
Pivmecillinam has high bioavailability (~70%), with 45% of the dose being secreted in urine as mecillinam within 6 h of administration [103]. Side effects are rare, with the most common being mild gastrointestinal symptoms [104]. The use of pivmecillinam as treatment for uncomplicated UTI is recommended by the European Society for Clinical Microbiology and Infectious Diseases, the European Association of Urology, and the Infectious Diseases Society of America [105]. Serum PK studies of 10 mg/kg mecillinam in healthy patients resulted in a mean Cmax of 61 mg/L (Table 2). There are no PK data from critically ill patients. The unbound fraction of the drug was calculated to be 90–95% [106]. The half-life of the drug was calculated to be 0.5 h.
Monte Carlo simulation of 5000 patients was performed with mean ± SD Cl 14.7 ± 1.4 L/h and VD 16.1 ± 2.8 L for 1 g q8h and 1 g q6h. As there are no PK/PD studies for mecillinam, the 50%T > MIC corresponding to 1-log kill against E. coli was used as found for most penicillins [57] (Table 3). The PTAs for the latter target were high (99%) for the tentative ECOFF of 0.5 mg/L for E. coli with the dosing regimen of 1 g q6h (Figure 5). PTAs are low for both dosing regimens for K. pneumoniae isolates as MIC90 (128 mg/L) is six-fold higher than E. coli according to EUCAST MIC distribution (Table 1). In the few patients where mecillinam was used against bacteremia, clinical and bacteriological success rates were 67% (10/15) and 87% (13/15), respectively [103,107]. Thus, mecillinam is a promising agent for treating gram-negative infections caused by E. coli at the dose of 1 g q6h.

8. Nitrofurantoin

Nitrofurantoin was introduced in clinical practice in 1953 [108] and is the most widely used antimicrobial within the nitrofuran class. Moreover, it is the only member of the nitrofuran family that is in use in human medicine and is available only as an oral formulation [109]. It is an old antibiotic that has been used for the treatment of uncomplicated UTI for decades, and its consumption increased as a first-line agent for the treatment of cystitis after the guidelines were updated in 2011 [105]. At low concentrations, nitrofurantoin inhibits the inducible synthesis of β-galactosidase and galactokinase without affecting total protein synthesis, while at higher concentrations, it inhibits enzymes of the citric acid cycle as well as DNA, RNA, and total protein synthesis in bacteria via a mechanism involving the reaction of electrophiles following bacterial reduction of nitrofurantoin with nucleophilic sites on bacterial macromolecules [110]. Concerning resistance mechanisms for nitrofurantoin, several mechanisms have been proposed, including mutations in nfsA and nfsB genes as well as the presence of the oqxAB gene [111]. Nitrofurantoin is mainly bacteriostatic but can also exhibit bactericidal effects when present at high concentrations (≥2 × MIC) [63,112]. Among the advantages of nitrofurantoin are the low prevalence of resistance amongst Enterobacterales and the low repercussions in commensal flora in comparison to the impact of quinolones or β-lactams [113,114]. Despite its extensive use, resistance rates are still low [115]. Its spectrum of activity includes ESBL-producing Enterobacterales—with the exception of Klebsiella and Proteae strains (e.g., Proteus, Morganella, and Providencia spp.), which show intrinsic resistance—Staphylococcus saprophyticus, and vancomycin-resistant enterococci [112,116,117]. Based on EUCAST MIC distribution, nitrofurantoin seems to be more potent against E. coli isolates, with an ECOFF of 64 mg/L (Table 1). Low susceptibility rates (MIC ≤ 32 mg/L) were found for K. pneumoniae (37.9%), P. aeruginosa (8%), and A. baumannii (8.3%) [118].
Following oral administration, nitrofurantoin is excreted rapidly via the kidney, resulting in high urine and low serum concentrations. The formulations of nitrofurantoin have been changed over the years, and currently, the clinical regimens of 100 mg q8h and 50 mg q6h are the most common. Furthermore, PK properties differ significantly between the products, and this is mainly because nitrofurantoin is not a uniform product because of different crystal sizes of nitrofurantoin [109].
PKs of nitrofurantoin were simulated in different dosing regimens in healthy female patients, as shown in Table 2, due to absence of data from critically ill patients. Clinical dosing regimens of 50 mg q6h and 100 mg q8h had resulted in AUC0–24 values of 4.43 ± 0.96 and 6.49 ± 2.9 mg·h/L and Cmax values of 0.326 ± 0.081 and 0.69 ± 0.35 mg/L, respectively. The unbound fraction of the drug was calculated to be between 25–50% [119]. The half-life of the drug ranged between 1.7–2.3 h.
The PK/PD index that best correlates with the drug’s antibacterial effect is still under investigation [120]. The PK/PD target of 82 %fT > MIC was associated with 3-log kill of E. coli isolates in an in vitro kinetic model [63], while no PK/PD study has been performed in animals (Table 3). Monte Carlo simulation of 5000 patients was performed with mean ± SD Cl 46.2 ± 18.6 L/h and VD 103.8 ± 65.9 L for a dosing regimen of 100 mg q8h and Cl 36.4 ± 11.4 L/h and VD 100 ± 49.6 L for 50 mg q8h [53]. The PTA for nitrofurantoin was low for E. coli with both dosing regimens (Figure 6). Oral nitrofurantoin has been used for UTI and pyelonephritis but not for the treatment of severe infections. Thus, oral nitrofurantoin is not promising for treating severe MDR gram-negative infections.

9. Minocycline

Minocycline is a semisynthetic second-generation tetracycline that was first introduced in clinical practice in the 1970s. The IV formulation of the drug was withdrawn from the US market in 2005 due to decreased use but was reintroduced in May 2009 as an important option for the treatment of MDR organisms [71]. Minocycline is bacteriostatic and inhibits protein synthesis by binding to the 30 s ribosomal subunit [121]. A variety of mechanisms, including modification or protection of the antibiotic target site and efflux pumps, are involved in bacterial resistance to minocycline, such as Tet(B) and RND- type efflux pumps [122]. Minocycline has a broad spectrum of action against aerobic and anaerobic gram-negative and gram-positive bacteria, including some strains of streptococci, staphylococci, and Haemophilus influenzae resistant to tetracycline. Minocycline is also currently approved by the FDA in the United States for the treatment of infections caused by susceptible A. baumannii isolates [64]. Based on EUCAST MIC distribution, minocycline is active against Enterobacterales with ECOFF values of 4 and 8 mg/L for E. coli and K. pneumoniae, respectively (Table 1). Moreover, recent surveillance studies have shown that minocycline is potent against MDR and carbapenem-resistant A. baumannii with MIC90 8 mg/L [71,123,124,125]. Regarding P. aeruginosa, a study demonstrated that 25% and 100% of isolates tested were inhibited by minocycline at a drug concentration of 25 and 50 mg/L, respectively [126].
Minocycline achieves excellent oral absorption and tissue penetration and a long elimination half-life, ranging from 15 to 23 h depending on administration of either 100 mg q12h or 200 mg q24h, respectively [127,128,129]. Most published PK data for IV minocycline concern healthy volunteers and are from studies conducted in 1970s [130]. Moreover, PK of minocycline has not been fully characterized in patients with creatinine clearance (CLCR) of <80 mL/min [129], and the FDA-approved product indicates that current data among patients with renal impairment are insufficient to determine if dose adjustments are warranted [130]. PK analysis of minocycline 200 mg resulted in an AUC0–24 of 24.3 ± 7.88 mg·h/L and a Cmax of 2.58 ± 1.33 mg/L (Table 2). The unbound fraction of minocycline in plasma was found to be 30 ± 12% [54]. The half-life of the drug was calculated to be 1.36 ± 0.45 h.
Monte Carlo simulation analysis of 5000 patients was performed with mean ± SD fAUC0–24 7.29 ± 2.36 mg·h/L for the dosing regimen tested. The PK/PD index that best correlates with the drug’s antibacterial effect seems to be fAUC/MIC, according to several studies [54,64,131,132], with an fAUC0–24/MIC 21.08 associated with 1-log kill of A. baumannii in pneumonia infection animal models [64] (Table 3). The PTAs for 200 mg q24h of minocycline were <4% at MIC90 of A. baumannii (Figure 7). Since there were no PK/PD targets for E. coli and K. pneumoniae, we used the same target as A. baumannii. The PTAs are expected to be low at ECOFFs of 4 mg/L for E. coli, 8 mg/L for K. pneumoniae, and MIC90 25–50 mg/L for P. aeruginosa. Successful treatment of MDR A. baumannii pneumonia with minocycline has been previously reported, although in most cases, minocycline was used in combination with other drugs and copathogens were involved [124]. Even with a higher exposure of fAUC0–24 of 25 mg·h/L previously reported [124], the PTA is high for isolates with MIC just up to 0.5 mg/L. Because human lung concentrations of minocycline are 4x plasma concentrations [128], if lung penetration in the rat model is lower, the PK/PD target must be adjusted. However, PTA will be still low even with higher exposure in lungs without covering all WT A. baumannii isolates (PTA 60% for MIC of 4 mg/L). Thus, minocycline is not a promising agent against MDR gram-negative infections.

10. Chloramphenicol

Chloramphenicol is the first broad-spectrum antibiotic to be manufactured synthetically on a large scale [133]. In most countries, it is available as topical agent and in some countries for parenteral administration. Nevertheless, due to rare but serious toxicity, it is now less used parenterally. Chloramphenicol binds to the 50S ribosomal subunit, inhibiting protein synthesis. Acquired chloramphenicol resistance comes from the production of the enzyme chloramphenicol acetyltransferase, but resistance can also be due to ribosomal modifications or altered permeability. It is active against various organisms, including gram-positive and gram-negative bacteria and anaerobes but less so against Bacteroides spp. Moreover, it is potent against Mycoplasma spp., Rickettsia spp., Chlamydia spp., and Leptospira spp. It is bacteriostatic but can be bactericidal at 2–4 × MIC against some gram-positive cocci, Neisseria spp., and Haemophilus influenzae [121]. Based on EUCAST MIC distribution, chloramphenicol’s ECOFF is 16 mg /L for E. coli, while the MIC90 was 8 mg/L for K. pneumoniae and ≥32 mg/L for MDR A. baumannii isolates (Table 1). Regarding other species, there is a study indicating that among MBL-positive isolates of P. aeruginosa, 68% were resistant to chloramphenicol according to CLSI guidelines [134]. Similar resistance rates (86.2%) were also found in a collection of MDR K. pneumoniae isolates carrying intI1 gene [135].
PKs of chloramphenicol were simulated in different dosing regimens and different groups of patients, as shown in Table 2. Briefly, clinical dosing regimens of mean 65.2 mg/kg/day and 1 g q6h resulted in AUC0–24 values of 468 ± 498 mg·h/L [55] and 518 mg·h/L [55], respectively, while single IV 30 mg/kg resulted in an AUC0–∞ of 72 ± 32 mg·h/L and a Cmax of 16.2 ± 9.1 mg/L [124]. The protein binding of chloramphenicol in plasma was found to be ~40% [136]. The half-life of the drug was calculated to be 1.2 ± 1.15 h.
PDs of chloramphenicol have never been studied in animal or human models. There is only one study that demonstrates the PDs of the related agent florfenicol, a derivative of chloramphenicol, indicating that AUC0–24/MIC is the PK/PD driver with an AUC24/MIC of 97.1 being associated with 1-log kill of E. coli in an ex vivo pig ileum model [65] (Table 3). Thus, Monte Carlo simulation analysis was performed with mean ± SD fAUC 187.2 ± 199.2 mg·h/L for a dosing regimen of 62.5 mg/kg/day, 28.8 ± 12.8 mg·h/L for 30 mg/kg/day and 207.2 ± 103.6 mg·h/L for 1 g q6h. The PTA was low at an ECOFF of 16 mg/L for E. coli for all dosing regimens (Figure 8). There are no clinical studies for the treatment of MDR gram-negative bacterial infections. Thus, chloramphenicol is not a promising agent.

11. Conclusions

In an era of increasing antibiotic resistance and scarcity of effective antimicrobials, there was a great interest on old antibiotics to combat MDR infections, as most of them demonstrated in vitro activity against these pathogens. Although they have been empirically used to treat infections—mainly mild infections but also more serious life-threatening infections in combination with other drugs—knowledge gaps on their PKs, PDs, and PK-PD characteristics and lack of clinical trials could not help to infer firm conclusion on their clinical efficacy [137]. Therefore, in recent years, the research community has made an enormous effort to fill in those gaps and explore the clinical potential of those antibiotics. In silico trials could help the efforts towards this direction, as modern tools of PK-PD analysis are utilized in order to determine whether old antibiotics could attain preclinical PK/PD targets in an approach used to assess new drugs during development and regulatory approval. The current review summarizes all in vitro MIC, PK, PK/PD, and PTA data in order to assess the clinical potential of old antibiotics to treat MDR infections and to provide hypotheses that need to be tested clinically. It has to be emphasized that in silico simulations conducted in the present analysis may not be extrapolated to other patient populations (e.g., critically ill, renal impairment, etc) as PKs may differ [138]. Furthermore, other factors, such as emergence of resistance, host factors, and infection sites where drugs may concentrate were not explored.
Among the old antibiotics tested, the most promising drugs were polymyxin B against E. coli, K. pneumoniae, and A. baumannii; temocillin against K. pneumoniae and E. coli; fosfomycin against E. coli and K. pneumoniae; and mecillinam against E. coli. As some MDR pathogens may have also high MICs to old antibiotics (polymyxin B and fosfomycin against CRE), the efficacy of the latter may be compromised against those pathogens. This emphasizes the importance of the careful use of old antibiotics with a clinical potential to treat MDR infections because irrational use can render them ineffective very quickly. The remaining antibiotics—colistin, minocycline, nitrofurantoin, and chloramphenicol—did not attain preclinical PK/PD targets in the current licensed forms and dosages (Table 4).
However, even for drug–bug pairs with low probability of PK/PD target attainment, old antibiotics may have a role in treating MDR infections in combination with other drugs, as multiple in vitro studies demonstrated the synergism of old antibiotics when combined with other antimicrobials. For example, synergistic or additive activity were observed in the combination of colistin with several other agents compared with any agent alone [139,140]. The potential synergistic activity of polymyxin B with other antibiotics has been evaluated in seven studies, most of them against A. baumannii isolates [70]. Considering temocillin, moderate synergism was found in combination with other beta-lactam antibiotics. Moreover, the combination of temocillin with fosfomycin demonstrated beneficial in vitro and in vivo results against E. coli strains that produced carbapenemases [141]. The emergence of fosfomycin resistance was prevented with the addition of temocillin, and the combination proved to be more bactericidal that fosfomycin alone [142]. Fosfomycin with even meropenem or imipenem can act synergistically against E. coli and A.baumannii strains in preventing the emergence of fosfomycin resistance [143,144]. The degree of synergism required for a clinically effective combination is unknown, although one could intuitively assume that a clinically effective combination would result in MIC reduction below the corresponding ECOFF or susceptibility breakpoint of the first drug at clinically achievable concentrations of the second drug and vice versa.
In conclusion, in silico modelling indicated clinical potentials of polymyxin B, temocillin, mecillinam, and fosfomycin against certain species of gram-negative MDR pathogens. Further studies are required in order to test the clinical efficacy of those antibiotics against MDR infections.

Author Contributions

Conception and supervision, J.M.; writing—original draft preparation, P.P.; writing—review and editing, J.M., S.P. and S.V. All authors have read and agreed to the published version of the manuscript.

Funding

This study received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest related to the present study.

References

  1. ECDC; WHO. Antimicrobial Resistance Surveillance in Europe; ECDC: Stockholm, Sweden, 2022; ISBN 9789294985521. [Google Scholar]
  2. Theuretzbacher, U.; Paul, M. Revival of old antibiotics: Structuring the re-development process to optimize usage. Clin. Microbiol. Infect. 2015, 21, 878–880. [Google Scholar] [CrossRef] [Green Version]
  3. Giske, C.G. Contemporary resistance trends and mechanisms for the old antibiotics colistin, temocillin, fosfomycin, mecillinam and nitrofurantoin. Clin. Microbiol. Infect. 2015, 21, 899–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Muller, A.E.; Theuretzbacher, U.; Mouton, J.W. Use of old antibiotics now and in the future from a pharmacokinetic/pharmacodynamic perspective. Clin. Microbiol. Infect. 2015, 21, 881–885. [Google Scholar] [CrossRef] [Green Version]
  5. Cassir, N.; Rolain, J.-M.; Brouqui, P. A new strategy to fight antimicrobial resistance: The revival of old antibiotics. Front. Microbiol. 2014, 5, 551. [Google Scholar] [CrossRef] [Green Version]
  6. Theuretzbacher, U.; Van Bambeke, F.; Cantó, R.; Giske, C.G.; Mouton, J.W.; Nation, R.L.; Paul, M.; Turnidge, J.D.; Kahlmeter, G. Reviving old antibiotics. J. Antimicrob. Chemother. 2015, 70, 2177–2181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Falagas, M.E.; Grammatikos, A.P.; Michalopoulos, A. Potential of old-generation antibiotics to address current need for new antibiotics. Expert Rev. Anti. Infect. Ther. 2008, 6, 593–600. [Google Scholar] [CrossRef]
  8. Committee for Medicinal Products for Human Use (CHMP). Guideline on the Use of Pharmacokinetics and Pharmacodynamics in the Development of Antibacterial Medicinal Products; European Medicines Agency: London, UK, 2016; pp. 1–17. [Google Scholar]
  9. Bulitta, J.B.; Hope, W.W.; Eakin, A.E.; Guina, T.; Tam, V.H.; Louie, A.; Drusano, G.L.; Hoover, J.L. Generating Robust and Informative Nonclinical In Vitro and In Vivo Bacterial Infection Model Efficacy Data To Support Translation to Humans. Antimicrob. Agents Chemother. 2019, 63, e02307-18. [Google Scholar] [CrossRef] [Green Version]
  10. Tsala, M.; Vourli, S.; Kotsakis, S.; Daikos, G.; Tzouvelekis, L.; Zerva, L.; Miriagou, V.; Meletiadis, J. Pharmacokinetic-pharmacodynamic modeling of meropenem against VIM producing Klebsiella pneumoniae isolates: Clinical implications. J. Med. Microbiol. 2015, 65, 211–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Mouton, J.W.; Dudley, M.N.; Cars, O.; Derendorf, H.; Drusano, G.L. Standardization of pharmacokinetic/pharmacodynamic (PK/PD) terminology for anti-infective drugs: An update. J. Antimicrob. Chemother. 2005, 55, 601–607. [Google Scholar] [CrossRef] [Green Version]
  12. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Colistin: Rationale for the EUCAST Clinical Breakpoints, version 1.0; EUCAST: Växjö, Sweden, 2010. [Google Scholar]
  13. Sader, H.S.; Farrell, D.J.; Castanheira, M.; Flamm, R.K.; Jones, R.N. Antimicrobial activity of ceftolozane/tazobactam tested against Pseudomonas aeruginosa and Enterobacteriaceae with various resistance patterns isolated in European hospitals (2011–2012). J. Antimicrob. Chemother. 2014, 69, 2713–2722. [Google Scholar] [CrossRef]
  14. Mikhail, S.; Singh, N.B.; Kebriaei, R.; Rice, S.A.; Stamper, K.C.; Castanheira, M.; Rybak, M.J. Evaluation of the Synergy of Ceftazidime-Avibactam in Combination with Meropenem, Amikacin, Aztreonam, Colistin, or Fosfomycin against Well-Characterized Multidrug-Resistant Klebsiella pneumoniae and Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2019, 63, e02233-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wang, Y.; Li, H.; Xie, X.; Wu, X.H.; Li, X.; Zhao, Z.; Luo, S.; Wan, Z.; Liu, J.; Fu, L.; et al. In vitro and in vivo assessment of the antibacterial activity of colistin alone and in combination with other antibiotics against Acinetobacter baumannii and Escherichia coli. J. Glob. Antimicrob. Resist. 2020, 20, 351–359. [Google Scholar] [CrossRef]
  16. Thet, K.T.; Lunha, K.; Srisrattakarn, A.; Lulitanond, A.; Tavichakorntrakool, R.; Kuwatjanakul, W.; Charoensri, N.; Chanawong, A. Colistin heteroresistance in carbapenem-resistant Acinetobacter baumannii clinical isolates from a Thai university hospital. World J. Microbiol. Biotechnol. 2020, 36, 102. [Google Scholar] [CrossRef]
  17. Gales, A.C.; Jones, R.N.; Sader, H.S. Contemporary activity of colistin and polymyxin B against a worldwide collection of Gram-negative pathogens: Results from the SENTRY Antimicrobial Surveillance Program (2006–2009). J. Antimicrob. Chemother. 2011, 66, 2070–2074. [Google Scholar] [CrossRef] [Green Version]
  18. Kuti, J.L.; Wang, Q.; Chen, H.; Li, H.; Wang, H.; Nicolau, D.P. Defining the potency of amikacin against Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii derived from Chinese hospitals using CLSI and inhalation-based breakpoints. Infect. Drug Resist. 2018, 11, 783. [Google Scholar] [CrossRef] [Green Version]
  19. Bratu, S.; Tolaney, P.; Karumudi, U.; Quale, J.; Mooty, M.; Nichani, S.; Landman, D. Carbapenemase-producing Klebsiella pneumoniae in Brooklyn, NY: Molecular epidemiology and in vitro activity of polymyxin B and other agents. J. Antimicrob. Chemother. 2005, 56, 128–132. [Google Scholar] [CrossRef]
  20. Cielo, N.C.; Belmonte, T.; Raro, O.H.F.; da Silva, R.M.C.; Wink, P.L.; Barth, A.L.; da Cunha, G.R.; Mott, M.P.; Riche, C.V.W.; Dias, C.; et al. Polymyxin B broth disk elution: A feasible and accurate methodology to determine polymyxin B susceptibility in Enterobacterales. Diagn. Microbiol. Infect. Dis. 2020, 98, 115099. [Google Scholar] [CrossRef]
  21. Gales, A.C.; Jones, R.N.; Sader, H.S. Global assessment of the antimicrobial activity of polymyxin B against 54,731 clinical isolates of Gram-negative bacilli: Report from the SENTRY antimicrobial surveillance programme (2001–2004). Clin. Microbiol. Infect. 2006, 12, 315–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hermes, D.M.; Pormann Pitt, C.; Lutz, L.; Teixeira, A.B.; Ribeiro, V.B.; Netto, B.; Martins, A.F.; Zavascki, A.P.; Barth, A.L. Evaluation of heteroresistance to polymyxin B among carbapenem-susceptible and -resistant Pseudomonas aeruginosa. J. Med. Microbiol. 2013, 62, 1184–1189. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, Y.; Zhao, C.; Wang, Q.; Wang, X.; Chen, H.; Li, H.; Zhang, F.; Wang, H. Evaluation of the in vitro activity of new polymyxin B analogue SPR206 against clinical MDR, colistin-resistant and tigecycline-resistant Gram-negative bacilli. J. Antimicrob. Chemother. 2020, 75, 2609–2615. [Google Scholar] [CrossRef] [PubMed]
  24. Zykov, I.N.; Sundsfjord, A.; Småbrekke, L.; Samuelsen, Ø. The antimicrobial activity of mecillinam, nitrofurantoin, temocillin and fosfomycin and comparative analysis of resistance patterns in a nationwide collection of ESBL-producing Escherichia coli in Norway 2010–2011. Infect. Dis. 2016, 48, 99–107. [Google Scholar] [CrossRef] [PubMed]
  25. Rodriguez-Villalobos, H.; Malaviolle, V.; Frankard, J.; de Mendonça, R.; Nonhoff, C.; Struelens, M.J. In vitro activity of temocillin against extended spectrum beta-lactamase-producing Escherichia coli. J. Antimicrob. Chemother. 2006, 57, 771–774. [Google Scholar] [CrossRef] [PubMed]
  26. Tärnberg, M.; Östholm-Balkhed, Å.; Monstein, H.J.; Hällgren, A.; Hanberger, H.; Nilsson, L.E. In vitro activity of beta-lactam antibiotics against CTX-M-producing Escherichia coli. Eur. J. Clin. Microbiol. Infect. Dis. 2011, 30, 981–987. [Google Scholar] [CrossRef] [PubMed]
  27. Livermore, D.M.; Hope, R.; Fagan, E.J.; Warner, M.; Woodford, N.; Potz, N. Activity of temocillin against prevalent ESBL- and AmpC-producing Enterobacteriaceae from south-east England. J. Antimicrob. Chemother. 2006, 57, 1012–1014. [Google Scholar] [CrossRef]
  28. Glupczynski, Y.; Huang, T.D.; Berhin, C.; Claeys, G.; Delmée, M.; Ide, L.; Ieven, G.; Pierard, D.; Rodriguez-Villalobos, H.; Struelens, M.; et al. In vitro activity of temocillin against prevalent extended-spectrum beta-lactamases producing Enterobacteriaceae from Belgian intensive care units. Eur. J. Clin. Microbiol. Infect. Dis. 2007, 26, 777–783. [Google Scholar] [CrossRef] [PubMed]
  29. Alexandre, K.; Fantin, B. Pharmacokinetics and Pharmacodynamics of Temocillin. Clin. Pharmacokinet. 2018, 57, 287–296. [Google Scholar] [CrossRef]
  30. Aprile, A.; Scalia, G.; Stefani, S.; Mezzatesta, M.L. In vitro fosfomycin study on concordance of susceptibility testing methods against ESBL and carbapenem-resistant Enterobacteriaceae. J. Glob. Antimicrob. Resist. 2020, 23, 286–289. [Google Scholar] [CrossRef]
  31. Kaase, M.; Szabados, F.; Anders, A.; Gatermann, S.G. Fosfomycin susceptibility in carbapenem-resistant Enterobacteriaceae from Germany. J. Clin. Microbiol. 2014, 52, 1893–1897. [Google Scholar] [CrossRef] [Green Version]
  32. Lai, B.; Zheng, B.; Li, Y.; Zhu, S.; Tong, Z. In vitro susceptibility of Escherichia coli strains isolated from urine samples obtained in mainland China to fosfomycin trometamol and other antibiotics: A 9-year surveillance study (2004–2012). BMC Infect. Dis. 2014, 14, 66. [Google Scholar] [CrossRef] [Green Version]
  33. Wootton, M.; Walsh, T.R.; Macfarlane, L.; Howe, R.A. Activity of mecillinam against Escherichia coli resistant to third-generation cephalosporins. J. Antimicrob. Chemother. 2010, 65, 79–81. [Google Scholar] [CrossRef]
  34. Marrs, E.C.L.; Day, K.M.; Perry, J.D. In vitro activity of mecillinam against Enterobacteriaceae with NDM-1 carbapenemase. J. Antimicrob. Chemother. 2014, 69, 2873–2875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Wentao, N.; Guobao, L.; Jin, Z.; Junchang, C.; Rui, W.; Zhancheng, G.; Youning, L. In vitro activity of minocycline combined with aminoglycosides against Klebsiella pneumoniae carbapenemase-producing K. pneumoniae. J. Antibiot. 2018, 71, 506–513. [Google Scholar] [CrossRef] [PubMed]
  36. Ni, W.; Li, G.; Zhao, J.; Cui, J.; Wang, R.; Gao, Z.; Liu, Y. Use of Monte Carlo simulation to evaluate the efficacy of tigecycline and minocycline for the treatment of pneumonia due to carbapenemase-producing Klebsiella pneumoniae. Infect. Dis. 2018, 50, 507–513. [Google Scholar] [CrossRef] [PubMed]
  37. Flamm, R.K.; Shortridge, D.; Castanheira, M.; Sader, H.S.; Pfaller, M.A. In Vitro Activity of Minocycline against U.S. Isolates of Acinetobacter baumannii-Acinetobacter calcoaceticus Species Complex, Stenotrophomonas maltophilia, and Burkholderia cepacia Complex: Results from the SENTRY Antimicrobial Surveillance Program, 2014. Antimicrob. Agents Chemother. 2019, 63, e01154-19. [Google Scholar] [CrossRef]
  38. Huang, L.Y.; Chen, T.L.; Lu, P.L.; Tsai, C.A.; Cho, W.L.; Chang, F.Y.; Fung, C.P.; Siu, L.K. Dissemination of multidrug-resistant, class 1 integron-carrying Acinetobacter baumannii isolates in Taiwan. Clin. Microbiol. Infect. 2008, 14, 1010–1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Tsala, M.; Vourli, S.; Georgiou, P.-C.; Pournaras, S.; Tsakris, A.; Daikos, G.L.; Mouton, J.W.; Meletiadis, J. Exploring colistin pharmacodynamics against Klebsiella pneumoniae: A need to revise current susceptibility breakpoints. J. Antimicrob. Chemother. 2018, 73, 953–961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Cheah, S.-E.; Wang, J.; Nguyen, V.T.T.; Turnidge, J.D.; Li, J.; Nation, R.L. New pharmacokinetic/pharmacodynamic studies of systemically administered colistin against Pseudomonas aeruginosa and Acinetobacter baumannii in mouse thigh and lung infection models: Smaller response in lung infection. J. Antimicrob. Chemother. 2015, 70, 3291–3297. [Google Scholar] [CrossRef] [Green Version]
  41. Ram, K.; Sheikh, S.; Bhati, R.K.; Tripathi, C.D.; Suri, J.C.; Meshram, G.G. Steady-state pharmacokinetic and pharmacodynamic profiling of colistin in critically ill patients with multi-drug–resistant gram-negative bacterial infections, along with differences in clinical, microbiological and safety outcome. Basic Clin. Pharmacol. Toxicol. 2021, 128, 128–140. [Google Scholar] [CrossRef]
  42. Plachouras, D.; Karvanen, M.; Friberg, L.E.; Papadomichelakis, E.; Antoniadou, A.; Tsangaris, I.; Karaiskos, I.; Poulakou, G.; Kontopidou, F.; Armaganidis, A.; et al. Population pharmacokinetic analysis of colistin methanesulfonate and colistin after intravenous administration in critically ill patients with infections caused by gram-negative bacteria. Antimicrob Agents Chemother 2009, 53, 3430–3436. [Google Scholar] [CrossRef] [Green Version]
  43. Daikos, G.L.; Skiada, A.; Pavleas, J.; Vafiadi, C.; Salatas, K.; Tofas, P.; Tzanetou, K.; Markogiannakis, A.; Thomopoulos, G.; Vafiadi, I.; et al. Serum bactericidal activity of three different dosing regimens of colistin with implications for optimum clinical use. J. Chemother. 2010, 22, 175–178. [Google Scholar] [CrossRef]
  44. Li, Y.; Deng, Y.; Zhu, Z.Y.; Liu, Y.P.; Xu, P.; Li, X.; Xie, Y.L.; Yao, H.C.; Yang, L.; Zhang, B.K.; et al. Population Pharmacokinetics of Polymyxin B and Dosage Optimization in Renal Transplant Patients. Front. Pharmacol. 2021, 12, 2184. [Google Scholar] [CrossRef] [PubMed]
  45. Manchandani, P.; Thamlikitkul, V.; Dubrovskaya, Y.; Babic, J.T.; Lye, D.C.; Lee, L.S.; Tam, V.H. Population Pharmacokinetics of Polymyxin B. Clin. Pharmacol. Ther. 2018, 104, 534–538. [Google Scholar] [CrossRef]
  46. Sandri, A.M.; Landersdorfer, C.B.; Jacob, J.; Boniatti, M.M.; Dalarosa, M.G.; Falci, D.R.; Behle, T.F.; Bordinhão, R.C.; Wang, J.; Forrest, A.; et al. Population pharmacokinetics of intravenous polymyxin B in critically ill patients: Implications for selection of dosage regimens. Clin. Infect. Dis. 2013, 57, 524–531. [Google Scholar] [CrossRef] [Green Version]
  47. De Joung, R.; Hens, R.; Basma, V.; Mouton, J.W.; Tulkens, P.M.; Carryn, S. Continuous versus intermittent infusion of temocillin, a directed spectrum penicillin for intensive care patients with nosocomial pneumonia: Stability, compatibility, population pharmacokinetic studies and breakpoint selection. J. Antimicrob. Chemother. 2008, 61, 382–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Laterre, P.F.; Wittebole, X.; Van De Velde, S.; Muller, A.E.; Mouton, J.W.; Carryn, S.; Tulkens, P.M.; Dugernier, T. Temocillin (6 g daily) in critically ill patients: Continuous infusion versus three times daily administration. J. Antimicrob. Chemother. 2015, 70, 891–898. [Google Scholar] [CrossRef]
  49. Hampel, B.; Feike, M.; Koeppe, P.; Lode, H. Pharmacokinetics of temocillin in volunteers. Drugs 1985, 29 (Suppl. 5), 99–102. [Google Scholar] [CrossRef]
  50. Merino-Bohórquez, V.; Docobo-Pérez, F.; Sojo, J.; Morales, I.; Lupión, C.; Martín, D.; Cameán, M.; Hope, W.; Pascual; Rodríguez-Baño, J. Population pharmacokinetics and pharmacodynamics of fosfomycin in non–critically ill patients with bacteremic urinary infection caused by multidrug-resistant Escherichia coli. Clin. Microbiol. Infect. 2018, 24, 1177–1183. [Google Scholar] [CrossRef] [Green Version]
  51. Roholt, K. Pharmacokinetic studies with mecillinam and pivmecillinam. J. Antimicrob. Chemother. 1977, 3, 71–81. [Google Scholar] [CrossRef]
  52. Gambertoglio, J.G.; Barriere, S.L.; Lin, E.T.; Conte, J.E. Pharmacokinetics of mecillinam in health subjects. Antimicrob. Agents Chemother. 1980, 18, 952–956. [Google Scholar] [CrossRef] [Green Version]
  53. Huttner, A.; Wijma, R.A.; Stewardson, A.J.; Olearo, F.; Von Dach, E.; Harbarth, S.; Brüggemann, R.J.M.; Mouton, J.W.; Muller, A.E. The pharmacokinetics of nitrofurantoin in healthy female volunteers: A randomized crossover study. J. Antimicrob. Chemother. 2019, 74, 1656–1661. [Google Scholar] [CrossRef]
  54. Lodise, T.P.; Van Wart, S.; Sund, Z.M.; Bressler, A.M.; Khan, A.; Makley, A.T.; Hamad, Y.; Salata, R.A.; Silveira, F.P.; Sims, M.D.; et al. Pharmacokinetic and Pharmacodynamic Profiling of Minocycline for Injection following a Single Infusion in Critically Ill Adults in a Phase IV Open-Label Multicenter Study (ACUMIN). Antimicrob. Agents Chemother. 2021, 65, e01809-20. [Google Scholar] [CrossRef]
  55. Slaughter, R.L.; Pieper, J.A.; Cerra, F.B.; Brodsky, B.; Koup, J.R. Chloramphenicol sodium succinate kinetics in critically ill patients. Clin. Pharmacol. Ther. 1980, 28, 69–77. [Google Scholar] [CrossRef] [PubMed]
  56. Acharya, G.P.; Davis, T.M.E.; Ho, M.; Harris, S.; Chataut, C.; Acharya, S.; Tuhladar, N.; Kafle, K.E.; Pokhrel, B.; Nosten, F.; et al. Factors affecting the pharmacokinetics of parenteral chloramphenicol in enteric fever. J. Antimicrob. Chemother. 1997, 40, 91–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Temocillin: Rationale for the EUCAST Clinical Breakpoints, version 1.0; EUCAST: Växjö, Sweden, 2010. [Google Scholar]
  58. Dudhani, R.V.; Turnidge, J.D.; Coulthard, K.; Milne, R.W.; Rayner, C.R.; Li, J.; Nation, R.L. Elucidation of the pharmacokinetic/pharmacodynamic determinant of colistin activity against Pseudomonas aeruginosa in murine thigh and lung infection models. Antimicrob. Agents Chemother. 2010, 54, 1117–1124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Van der Meijden, V.A.; Aranzana-Climent, H.; van der Spek, B.C.M.; de Winter, W.; Couet, J.; Meletiadis, A.E.; van den Muller, S.B. Pharmacokinetics and pharmodynamic properties of polymyxin B in murine infection models. 2022; Unpublished. [Google Scholar]
  60. Landersdorfer, C.B.; Wang, J.; Wirth, V.; Chen, K.; Kaye, K.S.; Tsuji, B.T.; Li, J.; Nation, R.L. Pharmacokinetics/pharmacodynamics of systemically administered polymyxin B against Klebsiella pneumoniae in mouse thigh and lung infection models. J. Antimicrob. Chemother. 2018, 73, 462–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Muller, A.E.; Raaphorst, M.; van der Meijden, A.; de Winter, B.C.M.; Meletiadis, J.; van den Muller, S.B. Pharmacodynamics of temocillin in neutropenic murine infection models. 2022, ahead of printing. 2022; ahead of printing. [Google Scholar]
  62. Lepak, A.J.; Zhao, M.; Vanscoy, B.; Taylor, D.S.; Ellis-Grosse, E.; Ambrose, P.G.; Andes, D.R. In Vivo Pharmacokinetics and Pharmacodynamics of ZTI-01 (Fosfomycin for Injection) in the Neutropenic Murine Thigh Infection Model against Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2017, 61, e00476-17. [Google Scholar] [CrossRef] [Green Version]
  63. Komp Lindgren, P.; Klockars, O.; Malmberg, C.; Cars, O. Pharmacodynamic studies of nitrofurantoin against common uropathogens. J. Antimicrob. Chemother. 2015, 70, 1076–1082. [Google Scholar] [CrossRef] [Green Version]
  64. Tarazi, Z.; Sabet, M.; Dudley, M.N.; Griffith, D.C. Pharmacodynamics of Minocycline against Acinetobacter baumannii in a Rat Pneumonia Model. Antimicrob. Agents Chemother. 2019, 63, e01671-18. [Google Scholar] [CrossRef] [Green Version]
  65. Lei, Z.; Liu, Q.; Khaliq, H.; Cao, J.; He, Q. Resistant cutoff values and optimal scheme establishments for florfenicol against Escherichia coli with PK-PD modeling analysis in pigs. J. Vet. Pharmacol. Ther. 2019, 42, 324–335. [Google Scholar] [CrossRef]
  66. Paul, M.; Daikos, G.L.; Durante-Mangoni, E.; Yahav, D.; Carmeli, Y.; Benattar, Y.D.; Skiada, A.; Andini, R.; Eliakim-Raz, N.; Nutman, A.; et al. Colistin alone versus colistin plus meropenem for treatment of severe infections caused by carbapenem-resistant Gram-negative bacteria: An open-label, randomised controlled trial. Lancet. Infect. Dis. 2018, 18, 391–400. [Google Scholar] [CrossRef]
  67. Nation, R.L.; Li, J.; Cars, O.; Couet, W.; Dudley, M.N.; Kaye, K.S.; Mouton, J.W.; Paterson, D.L.; Tam, V.H.; Theuretzbacher, U.; et al. Framework for optimisation of the clinical use of colistin and polymyxin B: The Prato polymyxin consensus. Lancet. Infect. Dis. 2015, 15, 225–234. [Google Scholar] [CrossRef] [PubMed]
  68. Trimble, M.J.; Mlynárčik, P.; Kolář, M.; Hancock, R.E.W. Polymyxin: Alternative Mechanisms of Action and Resistance. Cold Spring Harb. Perspect. Med. 2016, 6, a025288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Li, J.; Nation, R.L.; Milne, R.W.; Turnidge, J.D.; Coulthard, K. Evaluation of colistin as an agent against multi-resistant Gram-negative bacteria. Int. J. Antimicrob. Agents 2005, 25, 11–25. [Google Scholar] [CrossRef]
  70. Zavascki, A.P.; Goldani, L.Z.; Li, J.; Nation, R.L. Polymyxin B for the treatment of multidrug-resistant pathogens: A critical review. J. Antimicrob. Chemother. 2007, 60, 1206–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Bishburg, E.; Bishburg, K. Minocycline--an old drug for a new century: Emphasis on methicillin-resistant Staphylococcus aureus (MRSA) and Acinetobacter baumannii. Int. J. Antimicrob. Agents 2009, 34, 395–401. [Google Scholar] [CrossRef]
  72. Wang, P.; Zhang, Q.; Feng, M.; Sun, T.; Yang, J.; Zhang, X. Population Pharmacokinetics of Polymyxin B in Obese Patients for Resistant Gram-Negative Infections. Front. Pharmacol. 2021, 12, 754844. [Google Scholar] [CrossRef]
  73. Chen, N.; Guo, J.; Xie, J.; Xu, M.; Hao, X.; Ma, K.; Rao, Y. Population pharmacokinetics of polymyxin B: A systematic review. Ann. Transl. Med. 2022, 10, 231. [Google Scholar] [CrossRef]
  74. Kubin, C.J.; Nelson, B.C.; Miglis, C.; Scheetz, M.H.; Rhodes, N.J.; Avedissian, S.N.; Cremers, S.; Yin, M.T. Population Pharmacokinetics of Intravenous Polymyxin B from Clinical Samples. Antimicrob. Agents Chemother. 2018, 62, e01493-17. [Google Scholar] [CrossRef] [Green Version]
  75. Avedissian, S.N.; Liu, J.; Rhodes, N.J.; Lee, A.; Pais, G.M.; Hauser, A.R.; Scheetz, M.H. A Review of the Clinical Pharmacokinetics of Polymyxin B. Antibiotics 2019, 8, 31. [Google Scholar] [CrossRef] [Green Version]
  76. Kvitko, C.H.; Rigatto, M.H.; Moro, A.L.; Zavascki, A.P. Polymyxin B versus other antimicrobials for the treatment of pseudomonas aeruginosa bacteraemia. J. Antimicrob. Chemother. 2011, 66, 175–179. [Google Scholar] [CrossRef]
  77. Fang, J.; Li, H.; Zhang, M.; Shi, G.; Liu, M.; Wang, Y.; Bian, X. Efficacy of Ceftazidime-Avibactam Versus Polymyxin B and Risk Factors Affecting Clinical Outcomes in Patients With Carbapenem-Resistant Klebsiella pneumoniae Infections a Retrospective Study. Front. Pharmacol. 2021, 12, 780940. [Google Scholar] [CrossRef] [PubMed]
  78. Ouderkirk, J.P.; Nord, J.A.; Turett, G.S.; Kislak, J.W. Polymyxin B nephrotoxicity and efficacy against nosocomial infections caused by multiresistant gram-negative bacteria. Antimicrob. Agents Chemother. 2003, 47, 2659–2662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Rigatto, M.H.; Falci, D.R.; Zavascki, A.P. Clinical Use of Polymyxin B. Adv. Exp. Med. Biol. 2019, 1145, 197–218. [Google Scholar] [CrossRef] [PubMed]
  80. Livermore, D.M.; Tulkens, P.M. Temocillin revived. J. Antimicrob. Chemother. 2009, 63, 243–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Rodriguez-Villalobos, H.; Bogaerts, P.; Berhin, C.; Bauraing, C.; Deplano, A.; Montesinos, I.; de Mendonça, R.; Jans, B.; Glupczynski, Y. Trends in production of extended-spectrum beta-lactamases among Enterobacteriaceae of clinical interest: Results of a nationwide survey in Belgian hospitals. J. Antimicrob. Chemother. 2011, 66, 37–47. [Google Scholar] [CrossRef] [PubMed]
  82. Adams-Haduch, J.M.; Potoski, B.A.; Sidjabat, H.E.; Paterson, D.L.; Doi, Y. Activity of temocillin against KPC-producing Klebsiella pneumoniae and Escherichia coli. Antimicrob. Agents Chemother. 2009, 53, 2700–2701. [Google Scholar] [CrossRef] [Green Version]
  83. Chalhoub, H.; Pletzer, D.; Weingart, H.; Braun, Y.; Tunney, M.M.; Elborn, J.S.; Rodriguez-Villalobos, H.; Plésiat, P.; Kahl, B.C.; Denis, O.; et al. Mechanisms of intrinsic resistance and acquired susceptibility of Pseudomonas aeruginosa isolated from cystic fibrosis patients to temocillin, a revived antibiotic. Sci. Rep. 2017, 7, 40208. [Google Scholar] [CrossRef]
  84. Titelman, E.; Iversen, A.; Kahlmeter, G.; Giske, C.G. Antimicrobial susceptibility to parenteral and oral agents in a largely polyclonal collection of CTX-M-14 and CTX-M-15-producing Escherichia coli and Klebsiella pneumoniae. APMIS 2011, 119, 853–863. [Google Scholar] [CrossRef]
  85. Kim, B.; Kim, J.; Seo, M.R.; Wie, S.H.; Cho, Y.K.; Lim, S.K.; Lee, J.S.; Kwon, K.T.; Lee, H.; Cheong, H.J.; et al. Clinical characteristics of community-acquired acute pyelonephritis caused by ESBL-producing pathogens in South Korea. Infection 2013, 41, 603–612. [Google Scholar] [CrossRef]
  86. Balakrishnan, I.; Awad-El-Kariem, F.M.; Aali, A.; Kumari, P.; Mulla, R.; Tan, B.; Brudney, D.; Ladenheim, D.; Ghazy, A.; Khan, I.; et al. Temocillin use in England: Clinical and microbiological efficacies in infections caused by extended-spectrum and/or derepressed AmpC β-lactamase-producing Enterobacteriaceae. J. Antimicrob. Chemother. 2011, 66, 2628–2631. [Google Scholar] [CrossRef]
  87. Martinez-Beltran, J.; Loza, E.; Gomez-Alferez, A.; Romero-Vivas, J.; Bouza, E. Temocillin. In vitro activity compared with other antibiotics. Drugs 1985, 29 (Suppl. 5), 91–97. [Google Scholar] [CrossRef] [PubMed]
  88. Alexandre, K.; Leysour De Rohello, F.; Dahyot, S.; Etienne, M.; Tiret, I.; Gillibert, A.; Pestel-Caron, M.; Caron, F. Efficacy of temocillin against MDR Enterobacterales: A retrospective cohort study. J. Antimicrob. Chemother. 2021, 76, 784–788. [Google Scholar] [CrossRef] [PubMed]
  89. Falagas, M.E.; Athanasaki, F.; Voulgaris, G.L.; Triarides, N.A.; Vardakas, K.Z. Resistance to fosfomycin: Mechanisms, Frequency and Clinical Consequences. Int. J. Antimicrob. Agents 2019, 53, 22–28. [Google Scholar] [CrossRef] [PubMed]
  90. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Fosfomycin: Rationale for the EUCAST Clinical Breakpoints, version 1.0; EUCAST: Växjö, Sweden, 2022. [Google Scholar]
  91. Parker, S.; Lipman, J.; Koulenti, D.; Dimopoulos, G.; Roberts, J.A. What is the relevance of fosfomycin pharmacokinetics in the treatment of serious infections in critically ill patients? A systematic review. Int. J. Antimicrob. Agents 2013, 42, 289–293. [Google Scholar] [CrossRef] [PubMed]
  92. Huang, L.; Cao, M.; Hu, Y.; Zhang, R.; Xiao, Y.; Chen, G. Prevalence and mechanisms of fosfomycin resistance among KPC-producing Klebsiella pneumoniae clinical isolates in China. Int. J. Antimicrob. Agents 2021, 57, 106226. [Google Scholar] [CrossRef]
  93. Rodríguez-Gascón, A.; Canut-Blasco, A. Deciphering pharmacokinetics and pharmacodynamics of fosfomycin. Rev. Esp. Quimioter. 2019, 32, 19–24. [Google Scholar]
  94. Parker, S.L.; Frantzeskaki, F.; Wallis, S.C.; Diakaki, C.; Giamarellou, H.; Koulenti, D.; Karaiskos, I.; Lipman, J.; Dimopoulos, G.; Roberts, J.A. Population Pharmacokinetics of Fosfomycin in Critically Ill Patients. Antimicrob. Agents Chemother. 2015, 59, 6471–6476. [Google Scholar] [CrossRef] [Green Version]
  95. Kaye, K.S.; Rice, L.B.; Dane, A.L.; Stus, V.; Sagan, O.; Fedosiuk, E.; Das, A.F.; Skarinsky, D.; Eckburg, P.B.; Ellis-Grosse, E.J. Fosfomycin for Injection (ZTI-01) Versus Piperacillin-tazobactam for the Treatment of Complicated Urinary Tract Infection Including Acute Pyelonephritis: ZEUS, A Phase 2/3 Randomized Trial. Clin. Infect. Dis. 2019, 69, 2045–2056. [Google Scholar] [CrossRef] [Green Version]
  96. Nicolle, L.E. Pivmecillinam in the treatment of urinary tract infections. J. Antimicrob. Chemother. 2000, 46, 35–39. [Google Scholar] [CrossRef] [Green Version]
  97. Kerrn, M.B.; Frimodt-Møller, N.; Espersen, F. Urinary concentrations and urine ex-vivo effect of mecillinam and sulphamethizole. Clin. Microbiol. Infect. 2004, 10, 54–61. [Google Scholar] [CrossRef] [Green Version]
  98. Sullivan, Å.; Edlund, C.; Nord, C.E. Effect of antimicrobial agents on the ecological balance of human microflora. Lancet Infect. Dis. 2001, 1, 101–114. [Google Scholar] [CrossRef] [PubMed]
  99. Spratt, B.G. The mechanism of action of mecillinam. J. Antimicrob. Chemother. 1977, 3 (Suppl. B), 13–19. [Google Scholar] [CrossRef] [PubMed]
  100. Thulin, E.; Sundqvist, M.; Andersson, D.I. Amdinocillin (Mecillinam) resistance mutations in clinical isolates and laboratory-selected mutants of Escherichia coli. Antimicrob. Agents Chemother. 2015, 59, 1718–1727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Perea, E.J.; Palomares, J.C.; García-Iglesias, M.C. In vitro evaluation of new penicillins and cephalosporins upon P. aeruginosa and their interaction with mecillinam. Chemotherapy 1980, 26, 282–288. [Google Scholar] [CrossRef]
  102. Thabaut, A.; Durosoir, J.L.; Saliou, P. Comparative in vitro antibacterial activity of seven semi-synthetic penicillins against aerobic gram-negative bacteria and enterococci. Infection 1982, 10 (Suppl. 3), S249–S256. [Google Scholar] [CrossRef]
  103. Jansåker, F.; Frimodt-Møller, N.; Benfield, T.L.; Knudsen, J.D. Mecillinam for the treatment of acute pyelonephritis and bacteremia caused by Enterobacteriaceae: A literature review. Infect. Drug Resist. 2018, 11, 761–771. [Google Scholar] [CrossRef] [Green Version]
  104. Dewar, S.; Reed, L.C.; Koerner, R.J. Emerging clinical role of pivmecillinam in the treatment of urinary tract infection in the context of multidrug-resistant bacteria. J. Antimicrob. Chemother. 2014, 69, 303–308. [Google Scholar] [CrossRef] [Green Version]
  105. Gupta, K.; Hooton, T.M.; Naber, K.G.; Wullt, B.; Colgan, R.; Miller, L.G.; Moran, G.J.; Nicolle, L.E.; Raz, R.; Schaeffer, A.J.; et al. International clinical practice guidelines for the treatment of acute uncomplicated cystitis and pyelonephritis in women: A 2010 update by the Infectious Diseases Society of America and the European Society for Microbiology and Infectious Diseases. Clin. Infect. Dis. 2011, 52, e103–e120. [Google Scholar] [CrossRef] [Green Version]
  106. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Mecillinam: Rationale for the EUCAST Clinical Breakpoints, version 1.0; EUCAST: Växjö, Sweden, 2022. [Google Scholar]
  107. Neu, H.C. Amdinocillin: A novel penicillin. Antibacterial activity, pharmacology and clinical use. Pharmacotherapy 1985, 5, 1–10. [Google Scholar] [CrossRef]
  108. Shah, R.R.; Wade, G. Reappraisal of the risk/benefit of nitrofurantoin: Review of toxicity and efficacy. Advers. Drug React. Acute Poisoning Rev. 1989, 8, 183–201. [Google Scholar]
  109. Wijma, R.A.; Fransen, F.; Muller, A.E.; Mouton, J.W. Optimizing dosing of nitrofurantoin from a PK/PD point of view: What do we need to know? Drug Resist. Updat. 2019, 43, 1–9. [Google Scholar] [CrossRef] [PubMed]
  110. Mc Osker, C.C.; Fitzpatrick, P.M. Nitrofurantoin: Mechanism of action and implications for resistance development in common uropathogens. J. Antimicrob. Chemother. 1994, 33 (Suppl. A), 23–30. [Google Scholar] [CrossRef] [PubMed]
  111. Zhang, X.; Zhang, Y.; Wang, F.; Wang, C.; Chen, L.; Liu, H.; Lu, H.; Wen, H.; Zhou, T. Unravelling mechanisms of nitrofurantoin resistance and epidemiological characteristics among Escherichia coli clinical isolates. Int. J. Antimicrob. Agents 2018, 52, 226–232. [Google Scholar] [CrossRef] [PubMed]
  112. Fransen, F.; Melchers, M.J.B.; Meletiadis, J.; Mouton, J.W. Pharmacodynamics and differential activity of nitrofurantoin against ESBL-positive pathogens involved in urinary tract infections. J. Antimicrob. Chemother. 2016, 71, 2883–2889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Stewardson, A.J.; Vervoort, J.; Adriaenssens, N.; Coenen, S.; Godycki-Cwirko, M.; Kowalczyk, A.; Huttner, B.D.; Lammens, C.; Malhotra-Kumar, S.; Goossens, H.; et al. Effect of outpatient antibiotics for urinary tract infections on antimicrobial resistance among commensal Enterobacteriaceae: A multinational prospective cohort study. Clin. Microbiol. Infect. 2018, 24, 972–979. [Google Scholar] [CrossRef] [Green Version]
  114. Stewardson, A.J.; Gaïa, N.; François, P.; Malhotra-Kumar, S.; Delémont, C.; Martinez de Tejada, B.; Schrenzel, J.; Harbarth, S.; Lazarevic, V.; Vervoort, J.; et al. Collateral damage from oral ciprofloxacin versus nitrofurantoin in outpatients with urinary tract infections: A culture-free analysis of gut microbiota. Clin. Microbiol. Infect. 2015, 21, 344-e1. [Google Scholar] [CrossRef] [Green Version]
  115. De Greeff, S.C.; Mouton, J.W. NethMap 2018: Consumption of Antimicrobial Agents and Antimicrobial Resistance among Medically Important Bacteria in the Netherlands/MARAN 2018: Monitoring of Antimicrobial Resistance and Antibiotic Usage in Animals in the Netherlands in 2017; National Institute for Public Health and the Environment: Bilthoven, The Netherlands, 2018. [CrossRef]
  116. Mezzatesta, M.L.; La Rosa, G.; Maugeri, G.; Zingali, T.; Caio, C.; Novelli, A.; Stefani, S. In vitro activity of fosfomycin trometamol and other oral antibiotics against multidrug-resistant uropathogens. Int. J. Antimicrob. Agents 2017, 49, 763–766. [Google Scholar] [CrossRef]
  117. Zhanel, G.G.; Laing, N.M.; Nichol, K.A.; Palatnick, L.P.; Noreddin, A.; Hisanaga, T.; Johnson, J.L.; Hoban, D.J. Antibiotic activity against urinary tract infection (UTI) isolates of vancomycin-resistant enterococci (VRE): Results from the 2002 North American Vancomycin Resistant Enterococci Susceptibility Study (NAVRESS). J. Antimicrob. Chemother. 2003, 52, 382–388. [Google Scholar] [CrossRef] [Green Version]
  118. Alamri, A.; Hassan, B.; Hamid, M. Susceptibility of hospital-acquired uropathogens to first-line antimicrobial agents at a tertiary health-care hospital, Saudi Arabia. Urol. Ann. 2021, 13, 166. [Google Scholar] [CrossRef]
  119. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Nitrofurantoin: Rationale for the EUCAST Clinical Breakpoints, version 1.0; EUCAST: Växjö, Sweden, 2022. [Google Scholar]
  120. Wijma, R.A.; Huttner, A.; Koch, B.C.P.; Mouton, J.W.; Muller, A.E. Review of the pharmacokinetic properties of nitrofurantoin and nitroxoline. J. Antimicrob. Chemother. 2018, 73, 2916–2926. [Google Scholar] [CrossRef]
  121. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Minocycline: Rationale for the EUCAST Clinical Breakpoints, version 1.0; EUCAST: Växjö, Sweden, 2022. [Google Scholar]
  122. Asadi, A.; Abdi, M.; Kouhsari, E.; Panahi, P.; Sholeh, M.; Sadeghifard, N.; Amiriani, T.; Ahmadi, A.; Maleki, A.; Gholami, M. Minocycline, focus on mechanisms of resistance, antibacterial activity, and clinical effectiveness: Back to the future. J. Glob. Antimicrob. Resist. 2020, 22, 161–174. [Google Scholar] [CrossRef] [PubMed]
  123. Karageorgopoulos, D.E.; Falagas, M.E. Current control and treatment of multidrug-resistant Acinetobacter baumannii infections. Lancet. Infect. Dis. 2008, 8, 751–762. [Google Scholar] [CrossRef] [PubMed]
  124. Ritchie, D.J.; Garavaglia-Wilson, A. A review of intravenous minocycline for treatment of multidrug-resistant Acinetobacter infections. Clin. Infect. Dis. 2014, 59 (Suppl. 6), S374–S380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Scheetz, M.H.; Qi, C.; Warren, J.R.; Postelnick, M.J.; Zembower, T.; Obias, A.; Noskin, G.A. In vitro activities of various antimicrobials alone and in combination with tigecycline against carbapenem-intermediate or -resistant Acinetobacter baumannii. Antimicrob. Agents Chemother. 2007, 51, 1621–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Fedorko, J.; Katz, S.; Allnoch, H. In vitro activity of minocycline, a new tetracycline. Am. J. Med. Sci. 1968, 255, 252–258. [Google Scholar] [CrossRef]
  127. Agwuh, K.N.; MacGowan, A. Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines. J. Antimicrob. Chemother. 2006, 58, 256–265. [Google Scholar] [CrossRef] [Green Version]
  128. Watanabe, A.; Anzai, Y.; Niitsuma, K.; Saito, M.; Yanase, K.; Nakamura, M. Penetration of minocycline hydrochloride into lung tissue and sputum. Chemotherapy 2001, 47, 1–9. [Google Scholar] [CrossRef]
  129. Summary Product Information MINOCIN® Minocycline For Injection 100 Mg/Vial Intravenous. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/050444s047lbl.pdf (accessed on 14 October 2022).
  130. Welling, P.G.; Shaw, W.R.; Uman, S.J.; Tse, F.L.; Craig, W.A. Pharmacokinetics of minocycline in renal failure. Antimicrob. Agents Chemother. 1975, 8, 532–537. [Google Scholar] [CrossRef] [Green Version]
  131. Zhou, J.; Ledesma, K.R.; Chang, K.T.; Abodakpi, H.; Gao, S.; Tama, V.H. Pharmacokinetics and Pharmacodynamics of Minocycline against Acinetobacter baumannii in a Neutropenic Murine Pneumonia Model. Antimicrob. Agents Chemother. 2017, 61, e02371-16. [Google Scholar] [CrossRef] [Green Version]
  132. Alfouzan, W.A.; Noel, A.R.; Bowker, K.E.; Attwood, M.L.G.; Tomaselli, S.G.; MacGowan, A.P. Pharmacodynamics of minocycline against Acinetobacter baumannii studied in a pharmacokinetic model of infection. Int. J. Antimicrob. Agents 2017, 50, 715–717. [Google Scholar] [CrossRef]
  133. Ehrlich, J.; Bartz, Q.R.; Smith, R.M.; Joslyn, D.A.; Burkholder, P.R. Chloromycetin, a New Antibiotic From a Soil Actinomycete. Science 1947, 106, 417. [Google Scholar] [CrossRef]
  134. Rajput, A.; Saxena, R.; Singh, K.P.; Kumar, V.; Singh, S.; Gupta, A.; Singh, R.K. Prevalence and antibiotic resistance pattern of metallo-beta-lactamase-producing Pseudomonas aeruginosa from burn patients—experience of an Indian tertiary care hospital. J. Burn Care Res. 2010, 31, 264–268. [Google Scholar] [CrossRef]
  135. Ahangarzadeh Rezaee, M.; Langarizadeh, N.; Aghazadeh, M. First report of class 1 and class 2 integrons in multidrug-resistant Klebsiella pneumoniae isolates from northwest Iran. Jpn. J. Infect. Dis. 2012, 65, 256–259. [Google Scholar] [CrossRef] [Green Version]
  136. Burke, J.T.; Wargin, W.A.; Sherertz, R.J.; Sanders, K.L.; Blum, M.R.; Sarubbi, F.A. Pharmacokinetics of intravenous chloramphenicol sodium succinate in adult patients with normal renal and hepatic function. J. Pharmacokinet. Biopharm. 1982, 10, 601–614. [Google Scholar] [CrossRef]
  137. Zayyad, H.; Eliakim-Raz, N.; Leibovici, L.; Paul, M. Revival of old antibiotics: Needs, the state of evidence and expectations. Int. J. Antimicrob. Agents 2017, 49, 536–541. [Google Scholar] [CrossRef]
  138. Salem, A.H.; Zhanel, G.G.; Ibrahim, S.A.; Noreddin, A.M. Monte Carlo simulation analysis of ceftobiprole, dalbavancin, daptomycin, tigecycline, linezolid and vancomycin pharmacodynamics against intensive care unit-isolated methicillin-resistant Staphylococcus aureus. Clin. Exp. Pharmacol. Physiol. 2014, 41, 437–443. [Google Scholar] [CrossRef]
  139. Ontong, J.C.; Ozioma, N.F.; Voravuthikunchai Id, S.P.; Chusri Id, S. Synergistic antibacterial effects of colistin in combination with aminoglycoside, carbapenems, cephalosporins, fluoroquinolones, tetracyclines, fosfomycin, and piperacillin on multidrug resistant Klebsiella pneumoniae isolates. PLoS ONE 2021, 16, e0244673. [Google Scholar] [CrossRef]
  140. Tsala, M.; Vourli, S.; Georgiou, P.-C.; Pournaras, S.; Daikos, G.L.; Mouton, J.W.; Meletiadis, J. Triple combination of meropenem, colistin and tigecycline was bactericidal in a dynamic model despite mere additive interactions in chequerboard assays against carbapenemase-producing Klebsiella pneumoniae isolates. J. Antimicrob. Chemother. 2019, 74, 387–394. [Google Scholar] [CrossRef]
  141. Verbist, L.; Verhaegen, J. Effect of temocillin in combination with other beta-lactam antibiotics. Antimicrob. Agents Chemother. 1984, 25, 142–144. [Google Scholar] [CrossRef] [Green Version]
  142. Berleur, M.; Guérin, F.; Massias, L.; Chau, F.; Poujade, J.; Cattoir, V.; Fantin, B.; de Lastours, V. Activity of fosfomycin alone or combined with temocillin in vitro and in a murine model of peritonitis due to KPC-3- or OXA-48-producing Escherichia coli. J. Antimicrob. Chemother. 2018, 73, 3074–3080. [Google Scholar] [CrossRef]
  143. Singkham-in, U.; Chatsuwan, T. Synergism of imipenem with fosfomycin associated with the active cell wall recycling and heteroresistance in Acinetobacter calcoaceticus-baumannii complex. Sci. Rep. 2022, 12, 230. [Google Scholar] [CrossRef]
  144. Albiero, J.; Mazucheli, J.; Dos Reis Barros, J.P.; Dos Anjos Szczerepa, M.M.; Belini Nishiyama, S.A.; Carrara-Marroni, F.E.; Sy, S.; Fidler, M.; Sy, S.K.B.; Bronharo Tognim, M.C. Pharmacodynamic Attainment of the Synergism of Meropenem and Fosfomycin Combination against Pseudomonas aeruginosa Producing Metallo-β-Lactamase. Antimicrob. Agents Chemother. 2019, 63, e00126-19. [Google Scholar] [CrossRef]
Figure 1. Probability of target attainment (PTA) of colistin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Figure 1. Probability of target attainment (PTA) of colistin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Pharmaceuticals 15 01501 g001
Figure 2. Probability of target attainment (PTA) of polymyxin B at different dosing regimens against isolates with increasing MIC.
Figure 2. Probability of target attainment (PTA) of polymyxin B at different dosing regimens against isolates with increasing MIC.
Pharmaceuticals 15 01501 g002
Figure 3. Probability of target attainment (PTA) of temocillin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Figure 3. Probability of target attainment (PTA) of temocillin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Pharmaceuticals 15 01501 g003
Figure 4. Probability of target attainment (PTA) of fosfomycin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Figure 4. Probability of target attainment (PTA) of fosfomycin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Pharmaceuticals 15 01501 g004
Figure 5. Probability of target attainment (PTA) of mecillinam at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Figure 5. Probability of target attainment (PTA) of mecillinam at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Pharmaceuticals 15 01501 g005
Figure 6. Probability of target attainment (PTA) of nitrofurantoin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Figure 6. Probability of target attainment (PTA) of nitrofurantoin at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Pharmaceuticals 15 01501 g006
Figure 7. Probability of target attainment (PTA) of minocycline against isolates with increasing MIC. The cumulative fraction of response (CFR) for A. baumannii is shown for a collection of MDR isolates with MIC distribution by Flamm et al [37].
Figure 7. Probability of target attainment (PTA) of minocycline against isolates with increasing MIC. The cumulative fraction of response (CFR) for A. baumannii is shown for a collection of MDR isolates with MIC distribution by Flamm et al [37].
Pharmaceuticals 15 01501 g007
Figure 8. Probability of target attainment (PTA) of chloramphenicol at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Figure 8. Probability of target attainment (PTA) of chloramphenicol at different dosing regimens against isolates with increasing MIC. The cumulative fraction of response (CFR) is shown for a collection of isolates with the EUCAST MIC distribution.
Pharmaceuticals 15 01501 g008
Table 1. In vitro activity of old antibiotics against most common gram-negative bacteria.
Table 1. In vitro activity of old antibiotics against most common gram-negative bacteria.
Antimicrobial CompoundNo. of Isolates (Ref.)Mechanism bMIC Range (mg/L)MIC50
(mg/L)
MIC90
(mg/L)
ECOFF c
Colistin
E. coli6014 aNA2–1280.512
715 [13]ESBLNA≤0.250.5
K. pneumoniae1841 aNA0.125–5120.512
21 [14]CRKP0.25–2
633 [13]ESBLNA0.52
P. aeruginosa19,270 aNA0.06–128124
698 [13]MDRNA12
A. baumannii2879 aNA0.5–3212[2]
106 [15]NA0.125–320.51
75 [16]CRAB0.5–212
Polymyxin B
E. coli17,035 [17]NANA≤0.5≤0.5
86 [18]All including ESBL0.064–80.1250.25
K. pneumoniae96 [19]CRE0.5–1622
173 [20]CRE≤0.125–>64232
186 [18]All including ESBL0.064–40.250.5
P. aeruginosa8705 [21]NA≤1–>8≤12
124 [22]CRPsA includedNA12
A. baumannii18 [23]OXA-230.06–0.50.250.25
131 [18]All including CRAB0.125–>320.250.5
Temocillin
E. coli5702 aNA0.125–51283216
105 [24]ESBL2–32816
162 [25]ESBL1–64816
198 [26]CTX-M0.125–>128816
293 [27]CTX-M2–64816
40 [27]AmpC2-64816
K. pneumoniae605 aNA0.5–51222568
23 [28]Non-CTX-M ESBL4–321632
199 [27]CTX-M2–64832
P. aeruginosa104 [29]NA64–≥256≥256≥256
A. baumannii51 [29]NA2–≥256≥256≥256
Fosfomycin
E. coli2351 aNA0.125–512144
35 [30]ESBL≤0.5–1024132
24 [31]KPC, MBL,
OXA-48
≤0.25–2561256
528 [32]ESBLNANA2
K. pneumoniae1396 aNA0.5–51232256128
50 [30]KPC8–>204832>2048
27 [30]NDM, OXA-4816–>2048512>2048
50 [31]KPC, MBL,
OXA-48
0.5–>102416256
P. aeruginosa701 aNA1–51264128256
Mecillinam
E. coli1502 aNA0.03–5120.1252[0.5]
198 [26]CTX-M0.125–>12818
30 [33]Resistance to C3G 0.54
29 [34]NDM0.5–3248
K. pneumoniae175 aNA0.03–5120.25128
24 [34]NDM2–>328>32
Nitrofurantoin
E. coli4000 aNA1–256161664
105 [24]ESBL2–5121664
528 [32]ESBLNANA64
Minocycline
E. coli1498 aNA0.125–641164
K. pneumoniae938 aNA0.125–642168
70 [35]KPC0.06–64416
164 [36]CP-KpNA832
A. baumannii539 [37]MDR0.06–82>8
401 [37]XDR0.06–82>8
Chloramphenicol
E. coli45,852 aNA1–2564816
K. pneumoniae65 aNA1–25648
A. baumannii202 [38]MDR≤2–≥32≥32≥32
a Data from www.eucast.org, accessed on 23 October 2022. b Abbreviations: NA = Not Available information, mostly WT, MDR = Multiple Drug Resistance, ESBL = Extended Spectrum Beta-Lactamase, CRKP = Carbapenem-resistant K. pneumoniae, CRAB = Carbapenem-resistant Acinetobacter baumannii, CRE = carbapenem-resistant Enterobacterales, CRPsA = Carbapenem-resistant Pseudomonas aeruginosa, OXA = Oxacillinase, KPC = Klebsiella pneumoniae carbapenemase, MBL = Metallo-β-lactamase, NDM = New Delhi metallo beta lactamase, AmpC = Ampicillinase C. c Tentative ECOFF are in brackets. Cells are left empty when no data are available.
Table 2. Clinical pharmacokinetics of old antibiotics.
Table 2. Clinical pharmacokinetics of old antibiotics.
DrugDosage RegimenPatient Population (Ref.) CL
(L/h)
VD in L
(Mean ± SD)
%
Unbound
AUC0–24 (mg·h/L)
(Mean ± SD)
Cmax (mg/L)
(Mean ± SD)
t1/2 (h)
(Mean ± SD)
Colistin3 MU q8h13 ICU patients [43] 40%50.18 ± 10.743.34 ± 0.357.8 ± 0.76
4.5 MU q12h13 ICU patients [43] 40%60.71 ± 12.02.98 ± 0.278.8 ± 0.55
9 MU q24h13 ICU patients [43] 40%72.93 ± 38.575.83 ± 0.879.6 ± 0.62
Polymyxin B40–50 mg q12h 50 renal transplant patients [44]1.18 ± 0.112.09 ± 1.58 74.6 ± 17.818.15
119 ± 36.3 mg q12h–q24h35 adult patients [45]2.5 ± 1.134.3 ± 16.4 52.3 ± 14.8/
45.1 ± 17.3
10.1
0.45–3.38 mg/kg q12h–q24h24 critically ill patients [46]1.4V1 = 6.3, V2 = 23.14266.9 ± 21.62.79 ± 0.90 d11.9
Temocillin2g q12h10 ICU patients [47]2.44 ± 0.3914.3 ± 0.8723.7 ± 6.151856 ± 282147 ± 124.3 ± 0.3
2g q8h14 critically ill patients [48]3.69 ± 0.45V1 = 14 ± 2.51
V2 = 21.7 ± 4.52
411764170
0.5g10 healthy volunteers [49]1.5 ± 0.0910.5 ± 0.712344.1 ± 18.7 77.9 ± 28.45.2 ± 0.3
1 g10 healthy volunteers [49]1.78 ± 0.0811.9 ± 0.714573.3 ± 27.8160.8 ± 58.25.0 ± 0.2
2g10 healthy volunteers [49]2.62 ± 0.1616.8 ± 0.737784.5 ± 47.1236.1 ± 93.35.0 ± 0.2
Fosfomycin4g q6h16 non-critically ill [50]2.43 ± 1.6413.69 ± 2.81100 c5215.08 ± 1972.2422.6 ± 86.8
Mecillinam400 mg9 subjects [51] 90–95 c22 ± 528 ± 5
200 mg9 subjects [51] 90–95 c9.9 ± 1.512 ± 2
10 mg/kg12 healthy volunteers [52]14.7 ± 1.416.1 ± 2.890–95 c 610.85 ± 0.14
Nitrofurantoin
(oral)
50 mg q6h12 healthy adult female [53]36.4 ± 11.4100.0 ± 49.625–50 c4.43 ± 0.960.326 ± 0.0812.3 ± 1.8
100 mg q8h12 healthy adult female [53]46.2 ± 18.6103.8 ± 65.9 25–50 c6.49 ± 2.90.69 ± 0.351.7 ± 0.6
Minocycline200 mg55 critically ill patients [54]5.24 ± 2.63146 ± 5730 ± 1224.3 ± 7.882.58 ± 1.33T1/2,α = 1.36 ± 0.456
T1/2,β = 23.4 ± 9.53
Chloramphenicol
Sodium succinate
65.2 (32.3–114.4) mg/kg/day10 critically ill patients [55]21.24 ± 23.34 21 ± 8.4~40 *a468 ± 498 1.20 ± 1.15
30 mg/kg 7 patients [56]22.08 ± 10.32133 ± 56 34–63 *b72 ± 3216.2 ± 9.1
1 g q6h8 patients [57]7.72 ± 1.8723.1 ± 9.1~40 *a5188.4-26.00.57 ± 0.12
*a Based on Burke et al., J Pharmacokinet Biopharm. 1982, 10, 601–614. *b Based on DOI: 10.2165/00003088-198409030-00004. c Based on EUCAST rationale document. d Css,avg. Cells are left empty when no data are available.
Table 3. Plasma PK/PD target (mean ± SD) for old antibiotics.
Table 3. Plasma PK/PD target (mean ± SD) for old antibiotics.
DrugInfection Model
(Ref.)
DoseSpeciesPK/PD INDEXStasis1-Log Kill2-Log Kill3-Log Kill
ColistinThigh [58]5–160 mg/kg/day
q3h–q24h
P. aeruginosa (n = 3)
MIC = 0.5–1 mg/L
fAUC/MIC13.35 ± 4.5720.37 ± 4.1331.63 ± 4.2758.37 ± 7.27
Lung [58]5–160 mg/kg/day
q3h–q24h
P. aeruginosa (n = 3)
MIC = 0.5–1 mg/L
fAUC/MIC5.31 ± 1.1814.83 ± 2.3540.13 ± 5.01127 ± 19.29
In vitro [39]fCmax 9, 3 and 1.5 mg/L q8h–q24 hK. pneumoniae MDR(n = 2)
MIC = 0.5–2 mg/L
fAUC/MIC10 ± 2.114 ± 2.418 ± 3.124 ± 4.7
Polymyxin BThigh [59]4–512 mg/kg
q6h, q12h, q24h
E. coli MDR (n = 4)
MIC = 1 mg/L
fAUC/MIC63.5 ± 34.850.6 ± 3.8
Thigh [59]4–128 mg/kg q6h,
8–256 mg/kg q12h,
256– 512 mg/kg q24h
K. pneumoniae MDR (n = 5)
MIC = 0.5–2 mg/L
fAUC/MIC11.6 ± 22.139.7 ± 14.4
Thigh [60]0.5–120 mg/kg/dayK. pneumoniae (n = 3)
MIC = 0.25–1 mg/L
fAUC/MIC6.73 ± 6.2316.37 ± 12.17
TemocillinThigh [61]8–512 mg/kg q2h,
16–512 mg/kg q4h
E. coli ESBL (n = 4)
MIC = 8–16 mg/L
%fT > MIC66 ± 9.981.5 ± 14.4
K. pneumoniae ESBL (n = 4)
MIC = 8–64 mg/L
%fT > MIC63 ± 27.979 ± 6.4
Lung [61]16–1024 mg/kg q2h,
32–1024 mg/kg q4h
E. coli ESBL (n = 4)
MIC = 8–16 mg/L
%fT > MIC27.8 ± 13.835 ± 18.342.8 ± 23
K. pneumoniae ESBL (n = 4)
MIC = 8–64 mg/L
%fT > MIC35.8 ± 23.647.3 ± 21.4
FosfomycinThigh [62]12.5–6400 mg/kg/day q3h–q24hE. coli ESBL (n = 5)
MIC = 1–16 mg/L
fAUC0–24/MIC23.7 ± 15.398.9 ± 78.4
Thigh [62]12.5–6400 mg/kg/day
q3h–q24h
K. pneumoniae NDM (n = 3)
MIC = 4–16 mg/L
fAUC0–24/MIC11.1 ± 19.521.5 (n = 1)
Thigh [62]12.5–6400 mg/kg/day
q3h–q24h
P. aeruginosa (n = 2)
MIC = 8–16 mg/L
fAUC0–24/MIC14.6 ± 4.728.2 ± 17.82
MecillinamEUCAST RD Enterobacterales%fT > MIC30–35%~50%
NitrofurantoinIn vitro kinetic model [63]16 mg/LE. coli (n = 1)
MIC = 2 mg/L
%fT > MIC72% 82%
MinocyclineLung [64]0.46–180 mg/kg/day q12hA. baumannii (n = 6)
MIC = 0.03–4 mg/L
fAUC0–24/MIC13.75 ± 3.7621.08 ± 7.24
Florfenicol *Ex-vivo pig ileum [65]30 mg/kg
Single dose
E. coli (n = 1)
MIC = 8 mg/L
AUC0–24/MIC82.83 ± 3.5297.1 ± 4.12 101.6 ± 7.74
* Drug class of Chloramphenicol PK/PD target used. The PK/PD target of florfenicol was determined in the ileum fluid. Cells are left empty when no data are available or corresponding targets are not reached.
Table 4. Summary of old antibiotics with clinical potential against gram (-) isolates.
Table 4. Summary of old antibiotics with clinical potential against gram (-) isolates.
Old AntibioticsE. coliK. pneumoniaeP. aeruginosaA. baumannii
Colistin----
Polymyxin B-
Temocillin--
Fosfomycin--
Mecillinam---
Minocycline----
Nitrofurantoin----
Chloramphenicol----
✓ attain preclinical PKPD targets, - do not attain preclinical PKPD targets.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Paranos, P.; Vourli, S.; Pournaras, S.; Meletiadis, J. Assessing Clinical Potential of Old Antibiotics against Severe Infections by Multi-Drug-Resistant Gram-Negative Bacteria Using In Silico Modelling. Pharmaceuticals 2022, 15, 1501. https://doi.org/10.3390/ph15121501

AMA Style

Paranos P, Vourli S, Pournaras S, Meletiadis J. Assessing Clinical Potential of Old Antibiotics against Severe Infections by Multi-Drug-Resistant Gram-Negative Bacteria Using In Silico Modelling. Pharmaceuticals. 2022; 15(12):1501. https://doi.org/10.3390/ph15121501

Chicago/Turabian Style

Paranos, Paschalis, Sophia Vourli, Spyros Pournaras, and Joseph Meletiadis. 2022. "Assessing Clinical Potential of Old Antibiotics against Severe Infections by Multi-Drug-Resistant Gram-Negative Bacteria Using In Silico Modelling" Pharmaceuticals 15, no. 12: 1501. https://doi.org/10.3390/ph15121501

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