Next Article in Journal
Development of an NGS-Based Workflow for Improved Monitoring of Circulating Plasmids in Support of Risk Assessment of Antimicrobial Resistance Gene Dissemination
Next Article in Special Issue
New Antimicrobial Bioactivity against Multidrug-Resistant Gram-Positive Bacteria of Kinase Inhibitor IMD0354
Previous Article in Journal
Trends in Antimicrobial Susceptibility of Escherichia coli Isolates in a Taiwanese Child Cohort with Urinary Tract Infections between 2004 and 2018
Previous Article in Special Issue
Mersaquinone, A New Tetracene Derivative from the Marine-Derived Streptomyces sp. EG1 Exhibiting Activity against Methicillin-Resistant Staphylococcus aureus (MRSA)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fosfomycin as Partner Drug for Systemic Infection Management. A Systematic Review of Its Synergistic Properties from In Vitro and In Vivo Studies

by
Roberta Maria Antonello
1,
Luigi Principe
2,
Alberto Enrico Maraolo
3,
Valentina Viaggi
4,
Riccardo Pol
5,
Massimiliano Fabbiani
6,
Francesca Montagnani
6,7,
Antonio Lovecchio
1,
Roberto Luzzati
1 and
Stefano Di Bella
1,*
1
Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy
2
“San Giovanni di Dio” Hospital, 88900 Crotone, Italy
3
First Division of Infectious Diseases, Cotugno Hospital, AORN dei Colli, 80131 Naples, Italy
4
“A. Manzoni” Hospital, 23900 Lecco, Italy
5
Department of Infectious Diseases, Udine University, 33100 Udine, Italy
6
Department of Medical Sciences, Tropical and Infectious Diseases Unit, University Hospital of Siena, 53100 Siena, Italy
7
Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
*
Author to whom correspondence should be addressed.
Antibiotics 2020, 9(8), 500; https://doi.org/10.3390/antibiotics9080500
Submission received: 10 July 2020 / Revised: 1 August 2020 / Accepted: 3 August 2020 / Published: 10 August 2020
(This article belongs to the Special Issue Antibacterial Drug Discovery)

Abstract

:
Fosfomycin is being increasingly prescribed for multidrug-resistant bacterial infections. In patients with systemic involvement, intravenous fosfomycin is usually administered as a partner drug, as part of an antibiotic regimen. Hence, the knowledge of fosfomycin pharmacodynamic interactions (synergistic, additive, indifferent and antagonistic effect) is fundamental for a proper clinical management of severe bacterial infections. We performed a systematic review to point out fosfomycin’s synergistic properties, when administered with other antibiotics, in order to help clinicians to maximize drug efficacy optimizing its use in clinical practice. Interactions were more frequently additive or indifferent (65.4%). Synergism accounted for 33.7% of total interactions, while antagonism occurred sporadically (0.9%). Clinically significant synergistic interactions were mostly distributed in combination with penicillins (51%), carbapenems (43%), chloramphenicol (39%) and cephalosporins (33%) in Enterobactaerales; with linezolid (74%), tetracyclines (72%) and daptomycin (56%) in Staphylococcus aureus; with chloramphenicol (53%), aminoglycosides (43%) and cephalosporins (36%) against Pseudomonas aeruginosa; with daptomycin (97%) in Enterococcus spp. and with sulbactam (75%) and penicillins (60%) and in Acinetobacter spp. fosfomycin-based antibiotic associations benefit from increase in the bactericidal effect and prevention of antimicrobial resistances. Taken together, the presence of synergistic interactions and the nearly total absence of antagonisms, make fosfomycin a good partner drug in clinical practice.

Graphical Abstract

1. Introduction

Antimicrobial resistance (AMR) is a health issue of global concern, burdened with elevated costs and high morbidity and mortality rates. Limited therapeutic options and the increasing occurrence of resistance to last-resort antibiotics, i.e., colistin or carbapenems, make it necessary to reassess the role of “old” drugs while waiting for new antibiotics available on the market.
Fosfomycin (FOS) is an inhibitor of the synthesis of the bacterial wall acting with a unique mechanism of action. To carry out its action, FOS enters in the bacterial cell through the L-alpha-glycerophosphate and the hexose-6-phosphate transporter systems, interfering with the formation of the peptidoglycan precursor uridine diphosphate N-acetylmuramic acid (UDP-MurNAc) [1].
FOS, after being discovered in 1969 [2], has long been prescribed orally for low urinary tract infections (UTIs) and only recently has been repurposed, also intravenously and in combination, as a meropenem- and colistin-sparing agent to treat other infections (complicated UTIs, severe soft tissue infections, osteomyelitis, prostatitis, etc.) [1,3,4,5]. The excellent distribution in body sites, the safety and tolerability profile, as well as its affordability, make FOS a therapeutic option worth considering to treat multidrug-resistant (MDR) bacterial infections [6,7].
FOS is generally prescribed in association with at least another active agent. The association benefits from increase in the bactericidal effect of FOS, prevention of AMR, limitation of side effects thanks to lower dosages. Examples of commonly used empirical combination regimens including FOS are: Carbapenems + FOS, colistin + FOS, ceftolozane/tazobactam + FOS and tigecycline (TIG) + FOS.
We performed a systematic literature review concerning in vitro and in vivo studies to evaluate the synergistic effect of FOS in combination with other antibiotics and offer an overall view with clinically practical tables divided by antibiotic class.

2. Materials and Methods

This systematic review was carried out following the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA).
On 14 April 2020 we performed a MEDLINE/PubMed search using the search string “Fosfomycin"[Tw] AND (synerg*[Tw] OR association*[Tw] OR combin*[Tw] OR “together”[Tw] OR “additive”[Tw] OR “addition”[Tw] OR “checkerboard”[Tw] OR “chequerboard”[Tw] OR “time kill”[Tw] OR “time–kill”[Tw] OR “time–killing”[Tw] OR “time killing”[Tw])”.
1232 papers, from inception to 14 April 2020, were identified. Of these, 870 were excluded by title screening, 84 by abstract screening, 28 after full-text reading. Fifty-eight papers were excluded because written in a language different from English. 7 papers were excluded because full text was not available either online or in paper version. 185 papers were reviewed and discussed independently by seven authors (RMA, RP, AL, SDB, VV, LP, MF).
Common criteria for the evaluation of susceptibility and synergism were adopted by all authors.
Susceptibility. Susceptibility to FOS for Enterobacterales and Staphylococcus spp. was determined, according to the European Committee on Antimicrobial Susceptibility Testing (EUCAST) breakpoints, when the minimum inhibitory concentration (MIC) was ≤ 32 µg/mL. Enterococcus spp. were considered susceptible when exhibiting a MIC ≤ 64 µg/mL, according to the Clinical & Laboratory Standards Institute (CLSI) breakpoints. FOS breakpoints are not defined either by EUCAST or CLSI for Pseudomonas spp., Acinetobacter spp. and Streptococcus spp. Based on literature data, susceptibility was defined as a MIC ≤ 128 µg/mL for Pseudomonas spp. (ECOFF value), MIC ≤ 32 for Acinetobacter spp. and ≤64 µg/mL for Streptococcus spp. [8,9].
For all the antibiotics tested in combination, EUCAST breakpoints was considered at first and CLSI breakpoints were considered when EUCAST breakpoints were not available. Breakpoints adopted are specified in each paragraph.
Synergistic effect. Checkerboard assay: fractional inhibitory concentration index (FICI) ≤ 0.5. FICI is defined as follows:
F I C I = M I C   F O S   i n   c o m b i n a t i o n M I C   F O S   a l o n e + M I C   o t h e r   a n t i b i o t i c   i n   c o m b i n a t i o n M I C   o t h e r   a n t i b i o t i c   a l o n e
.
Time–kill assay: ratio of effective concentrations concordant with FICI or ≥2 log kill.
Additive effect. Checkerboard assay: 0.5 < FICI ≤ 1. Time–kill assay: ratio of effective concentrations concordant with FICI or 1 < log kill < 2.
Indifferent effect. Checkerboard assay: 1 < FICI < 4. Time–kill assay: ratio of effective concentrations concordant with FICI or ± 1 log kill.
Antagonistic effect. Checkerboard assay: FICI ≥ 4. Time–kill assay: ratio of effective concentrations concordant with FICI or < 1 log kill.
For in vitro studies using a method different from checkerboard or time–kill assay, or in case data on effective concentrations were not available, synergism was evaluated according to the authors’ judgment.
For studies performed in vivo, synergism was established with the same ratio of effective concentrations considered for checkerboard assays or with the same log kill considered for time–kill assays. When these data were not reported in the paper, synergism was evaluated according to the authors’ judgment.

3. Results

For a better comprehension, a table with reviewed papers and a summary of most relevant results is proposed for each antibiotic class.

3.1. Penicillins

Twenty-eight papers evaluating FOS in combination with penicillins, penicillins + β-lactamase inhibitors, penicillinase-resistant penicillins were reviewed (Table 1). Breakpoints for penicillins were inferred from EUCAST breakpoints [10]. Penicillins are β-lactam antibiotics that acts through the inhibition of enzymes needed for peptidoglycans cross linking. Effect of FOS in combination with penicillins varied greatly according with the bacterial species considered. The highest rates of synergistic effect were observed against Enterobacterales and Acinetobacter spp. Despite this, Avery et al. [11] reported high rates of indifferent effect of FOS + piperacillin/tazobactam (PIP/TAZ) against PIP/TAZ-resistant Enterobacterales. Antagonistic effect was observed against one isolate of S. aureus with the combination FOS + methicillin [12] and against 6 biofilm-producer Enterococcus faecalis isolates with the combination FOS + ampicillin [13]. Four studies [14,15,16,17] performed in vivo experiments, with no substantial differences in results when compared with results obtained in vitro.
The combination of penicillin + FOS retains additive/synergistic effects against ~50% of Enterobacterales, Acinetobacter spp., Staphylococcus spp., and Streptococcus spp. strains.

3.2. Cephalosporins

Forty-one papers evaluating FOS in combination with cephalosporins and cephalosporins + β-lactamase inhibitors were reviewed (Table 2). Breakpoints for cephalosporins were inferred from EUCAST breakpoints [10]. Cephalosporins are β-lactam antibiotics that acts disrupting the peptidoglycan synthesis like penicillins, but are less susceptible to β-lactamases. Some studies reported discordant results on the effect of FOS in combination with a cephalosporin against clinical isolates, particularly against Staphylococcus spp. [18,19,20] and Enterobacterales isolates [11,14,21]. Antagonistic effect was observed against 4 Pseudomonas aeruginosa isolates with the combination FOS + ceftazidime [22], 1 S. aureus and 1 Staphylococcus epidermidis isolates with the combination FOS + ceftriaxone [19]. 9 in vivo studies [17,23,24,25,26,27,28,29,30] performed with different strains (Escherichia coli, P. aeruginosa, S. aureus, Streptococcus pneumoniae, Streptococcus sanguis) confirmed results obtained in vitro or resulted in higher synergistic effect (additive effect only against 3 S. aureus isolates [25,26]).
Cephalosporins + β-lactamase inhibitors, often chosen by clinicians to treat MDR infections, resulted in moderate rates of synergistic effect in combination with FOS. Against Enterobacterales, the combination ceftolozane/tazobactam + FOS resulted synergistic in 16.3% of cases (49 isolates tested [11]), while the combination ceftazidime/avibactam + FOS was synergistic in 28.8% of cases (66 isolates tested [11,21,31]). Against P. aeruginosa, the combination ceftolozane/tazobactam + FOS resulted synergistic in 71.1% of cases (45 isolates tested [32,33,34]), while the combination ceftazidime/avibactam + FOS was synergistic in 31.6% of cases (38 isolates tested [21,29,33]).
The combination of cephalosporins or cephalosporins + β-lactamase inhibitors + FOS appears to be clinically appealing especially against infections sustained by Enterobacterales and Pseudomonas spp.

3.3. Carbapenems

Forty-four papers evaluating FOS in combination with carbapenems were reviewed (Table 3). Carbapenems are β-lactam antibiotics that inhibit bacterial cell wall synthesis by binding to penicillin-binding proteins. Carbapenems are β-lactams “last-resort” used intravenously to treat severe infections. Imipenem (IMI) breakpoints are ≤ 2 µg/mL for Enterobacterales, Acinetobacter spp., S. pneumoniae and ≤ 0.001 µg/mL for Pseudomonas spp. and Staphylococcus spp. Meropenem breakpoints are ≤2 µg/mL for Enterobacterales, Acinetobacter spp., Pseudomonas spp., S. pneumoniae and ≤4 µg/mL for Staphylococcus spp. Ertapenem (ERT) breakpoints are ≤0.5 µg/mL for Enterobacterales, S. pneumoniae and ≤4 µg/mL for Staphylococcus spp. [10].
Synergism rates were not unanimous on all studies, but antagonistic effect was observed only in 2 isolates of P. aeruginosa in the study by Pruekprasert et al. [22] and in 1 isolate of S. aureus in the study by Quentin et al. [35]. No evident differences in the synergistic effect was observed depending on the carbapenem tested. The association FOS + carbapenem often resulted, when reported, in FOS- and/or carbapenem-susceptibility restoration. Three authors performed in vivo experiments using methicillin-resistant Staphylococcus aureus (MRSA) isolates: in two studies [28,36] the results in vivo were concordant with those found in vitro, while in the third study the combination in vivo resulted less effective [37].
From the clinical point of view the combination of carbapenems + FOS against Enterobacterales, P. aeruginosa end Acinetobacter spp. appears appealing.

3.4. Monobactams

Five papers evaluating FOS in combination with aztreonam (ATM) were reviewed (Table 4). ATM is a synthetic antibiotic whose susceptibility is often preserved also in those strains which are resistant to other β-lactam antibiotics. The mechanism of action is similar to penicillins. ATM breakpoints are ≤1 µg/mL for Enterobacterales and ≤0.001 µg/mL for Pseudomonas spp. [10].
The largest study evaluating FOS in combination with ATM on Enterobacterales isolates [33] reported an indifferent effect on most (64.6%) isolates. The combination was reported to have an additive effect on most isolates of P. aeruginosa [33,38], sometimes leading to ATM susceptibility restoration [33,39]. There were no in vivo studies evaluating this combination.

3.5. Quinolones

Twenty-nine papers evaluating FOS in combination with quinolones were reviewed (Table 5). Quinolones are bactericidal antibiotics that directly inhibit bacterial DNA synthesis. Breakpoints for quinolones were inferred from EUCAST breakpoints [10]. Synergism rates were not unanimous on all studies for isolates of P. aeruginosa. In 1 in vivo study synergism rate was 100% according to Mikuniya et al. [40]. Antagonism was observed in 1 in vivo [41] and 1 in vitro studies [39]. For E. coli isolates there was a weak synergism. In a recent in vitro study there was complete FOS and ciprofloxacin susceptibility restoration [42]. The combinations showed different synergistic rates for Staphylococcus spp. isolates with 100% synergistic rate in 1 in vitro study [43] and in 1 in vivo study [44]. No antagonism was observed for E. coli and Staphylococcus spp. isolates. There were some differences in the synergistic effect depending on the quinolone tested. The most frequent effect of FOS + ciprofloxacin was indifferent even though it showed in vitro 95% synergistic effect with S. aureus [45] The combination with levofloxacin showed mainly an additive effect in P. aeruginosa [38,39,46] and in Acinetobacter spp. [38] isolates.
In summary good additive/synergistic effect rates are reported when quinolones + FOS are used against S. aureus and P. aeruginosa isolates.

3.6. Aminoglycosides

Aminoglycosides (AMG) act through inhibition of protein synthesis, resulting in a potent and broad-spectrum antibacterial activity but with a potential high nephro- and oto-toxicity [47]. In the attempt to overcome increasing aminoglycosides resistance, development of novel AMG (such as arbekacin and plazomicin) has occurred, but combination strategies are important opportunities to treat resistant bacteria and to reduce toxicity. Inhaled delivery of tobramycin, allowing for greater exposure within the lungs and reducing systemic toxicity, is also approved for the treatment of patients with chronic P. aeruginosa lung infection associated with cystic fibrosis (CF) in United States and Europe [47]. Overall, 41 papers evaluating FOS in combinations with AMG were reviewed (Table 6). Available EUCAST aminoglycosides breakpoints were applied in all studies except one [48]. Due to the peculiarity of possible AMG therapeutic use (e.g. inhaled formulation in cystic fibrosis), many studies investigated the AMG + FOS combination also when administered by inhaled topical use; moreover, the activity of this combination on biofilm formation and in anaerobic conditions was also evaluated. Different AMG were tested as partner of FOS towards several bacterial species in a total of 67 evaluations: mainly gentamicin (31.3%, n = 21), amikacin (23.9%, n = 16) and tobramycin (22.4%, n = 15) were used. Synergism rates were not unanimous on all studies, considering the different bacteria analyzed and the different types of aminoglycosides tested. Overall, a synergistic effect of FOS together with different AMG, even if with different percentages, was revealed in 51 evaluations (74.6%). No synergism was reported in 16 cases (23.9%), even regarding effects on P. aeruginosa and Acinetobacter spp. In one study, data on synergism were not available [49]: however, a potential beneficial effect was indeed reported, demonstrating that FOS enhanced the activity of tobramycin with a 100% additive effect during in vitro evaluation on P. aeruginosa biofilms on cystic fibrosis airway epithelial cells. An antagonistic effect, testing the combination of FOS with gentamicin, was reported in 1985 by Alvarez et al. in 2.7% of 148 MRSA isolates [12] and in 2005 by Pruekprasert et al. in 27% of 22 P. aeruginosa strains [22].
Focusing on different bacterial strains, generally a synergistic or additive effect of FOS + AMG was demonstrated on KPC-producing K. pneumoniae [50,51,52]; however, Souli et al. observed an indifferent effect of FOS + gentamycin combination in all of their tested KPC+ strains [53].
When tested, a generally positive effect of FOS and AMG combination on biofilm formation and an improved AMG activity in anaerobic conditions were also reported for P. aeruginosa and Acinetobacter spp., resulting moreover in lower required AMG doses.
Activity of FOS plus an AMG was also evaluated against Streptococcus spp. (streptomycin) and Neisseria gonorrhoeae (both, gentamicin) in two studies [14,54]: No synergistic effect was revealed but antagonism was not even reported. Interestingly, synergistic activity (assessed as a fourfold reduction of MIC when fosfomycin was combined with gentamicin 1 mcg/mL) and additive effect were revealed for 8 vancomycin-resistant E. faecium (VRE) isolates (63% and 13%, respectively) [55].
The combination of AMG + FOS against P. aeruginosa appears to be the most clinically appealing.

3.7. Macrolides

Six papers evaluating FOS in combination with macrolides, in particular with erythromycin (ERY), azithromycin (AZT), clarithromycin (CLT), or midecamycin (MDM), were reviewed (Table 7). Macrolides are a large class of antibiotics that act binding 50S ribosomal subunit, inhibiting bacterial proteins synthesis. They have broad-spectrum activity, mainly against many Gram-positive bacteria and some Gram-negative bacteria [56]. Only one in vitro study evaluated FOS + ERY combination against Enterobacterales (87 strains of E. cloacae, E. coli, Proteus spp. and Klebsiella pneumoniae), reporting synergistic effect against 52% of isolates and additive effect against 30% [14]; in the same study FOS + ERY combination was also tested against P. aeruginosa and S. aureus, proving in most cases additive effect or, less frequently, synergistic effect [14]. When this combination was tested against Streptococcus spp. synergistic effect was observed against 15% of isolates, while additive (27%) or indifferent (58%) was seen against the remaining [14]. Some studies evaluated FOS + AZT combination, reporting indifferent effect in 100% of cases, either when tested against N. gonorrhoeae (2 studies) [54,57] or against S. epidermidis (1 study) [58]. Finally, FOS + CLT and FOS + MDM combinations were evaluated against S. pseudointermedius and P. aeruginosa respectively; in both cases additive or synergistic effect was demonstrated in vitro or in vivo experiments [59,60]. No antagonistic effect was observed for any combination against any isolate.
From the clinical point of view the combination of macrolides + FOS appears the less appealing.

3.8. Glycopeptides

Eighteen articles evaluating FOS in combination with glycopeptides (vancomycin and teicoplanin) have been reviewed (Table 8). Articles were from Spain (n = 5), Taiwan (n = 3), China (n = 2), France (n = 2), Germany (n = 2), Italy (n = 2), Austria (n = 1), and Brazil (n = 1).
Glycopeptides possess an antimicrobial activity selectively directed against Gram-positive bacteria, while Gram-negatives are protected by the outer membrane that is impermeable to these antibiotics. Glycopeptides inhibit the peptidoglycan synthesis by interacting with the terminal D-alanyl-D-alanine present on the pentapeptide side chains of the peptidoglycan precursors.
384 strains have been studied, belonging to several species as S. aureus (n = 219), S. epidermidis (n = 52), E. faecalis (n = 39), S. pneumoniae (n = 28), Acinetobacter baumannii (n = 20), Enterococcus faecium (n = 16) and other coagulase-negative staphylococci (CoNS) (n = 10). Synergy was detected with FOS-vancomycin (VAN) combination (40 out of 308 strains tested, 13%) in 33.3% of E. faecalis, 30% of E. faecium, 16.7% of S. aureus, 13.5% of S. epidermidis, and 3.6% of S. pneumoniae. Higher rates of synergistic interactions were detected with FOS-teicoplanin (TEC) combination (63 out of 130 strains tested, 48.5%) in 71.8% of E. faecalis, 43.7% of E. faecium, 60% of other CoNS, 34.3% of S. aureus and 33.3% S. epidermidis. Synergistic concentration ranges were 1-64 mg/L for FOS, 1-7.5 mg/L for VAN and only 8 mg/L for TEC. Regarding resistant isolates, FOS-VAN synergy was detected in one heterogeneous glycopeptide-intermediate Staphylococcus aureus (hGISA), 27 MRSA, 5 S. aureus strains with borderline MIC values for VAN (2 mg/L) and in 6 VRE strains, while FOS-TEC in 10 MRSA and 11 VRE strains. Antagonism FOS-VAN was detected in 5 S. aureus and one S. epidermidis strains. Only in 8 FOS-resistant S. aureus strains the activity of FOS was restored in combination with VAN. In vivo application of FOS-VAN combinations showed significant survival of ≥50% of treated animals or patients with infections caused by S. aureus or S. epidermidis [24,36,61,62,63].
In summary the combination of VAN + FOS resulted in good synergistic effect rates against Enterococcus spp. isolates and seems to be the most clinically relevant combination.

3.9. Tetracyclines

Ten papers evaluating FOS in combination with tetracyclines, mostly with minocycline (MIN) and in few cases with doxycycline (DOX) or tetracycline (TEC), were reviewed (Table 9). Tetracyclines are a large class of antibiotics that acts binding the 30S ribosomal subunits, inhibiting bacterial proteins synthesis. They have broad-spectrum activity, being active against many Gram-positive bacteria, Gram-negative, and atypical bacteria [64]. Almost all studies evaluated in vitro FOS + MIN combination against different bacterial species. When evaluated against Enterobacterales (20 strains), FOS + MIN proved to have additive effect most of the time (65% of isolate), but only in few cases synergistic effect [38]. Similar results were observed when it was tested against multidrug-resistant P. aeruginosa [38] and A. baumannii isolates; furthermore, in the last case, complete restoration of susceptibility of MIN was reported [65]. Only one study evaluated FOS + TEC combination against Enterobacterales (100 isolates), observing indifference in almost 100% of cases [66]. 2 studies evaluated FOS + MIN combination against vancomycin-resistant E. faecium or E. faecalis (51 strains), reporting most often indifferent effect and some sporadic case of synergism [13,67]. Otherwise, FOS + DOX combination was tested once against 24 isolates of vancomycin-resistant E. faecium, demonstrating to have synergistic or additive effect in most of cases [68]. Finally, when FOS + MIN was tested against MRSA (152, strains, 3 studies) proved to have synergistic effect in numerous cases [18,69,70]. No study reported any case of antagonism.
The combination of minocycline + FOS against A. baumannii appears interesting.

3.10. Polymyxins

Thirty-two papers evaluating FOS in combination with polymyxins were reviewed (Table 10). Polymyxins are bactericidal drugs that bind to lipopolysaccharide (LPS) and phospholipids in the outer cell membrane of Gram-negative bacteria and leads to disruption of this. Twenty-eight papers evaluated colistin. Colistin breakpoints are ≤ 2 µg/mL for Enterobacterales, Acinetobacter spp. and Pseudomonas spp. according to the EUCAST [10]. Synergism rates were not unanimous on all studies but was reported in 23/29 papers. Synergisms rate were 100% in 2 in vitro studies against K. pneumoniae [50,71] and 2 in vivo studies respectively against A. baumannii and E.coli [72,73]. The overall effect was indifferent on most isolates of P. aeruginosa and Enterobacterales. Antagonism was reported in vitro against K. pneumoniae and A. baumannii. In particular the combination was antagonist in 100% of all K. pneumoniae OXA-48 isolates according to Evren et al. [74].
Four papers evaluated polymyxin B. Polymyxin B breakpoints for Enterobacterales, Acinetobacter spp. and Pseudomonas spp. are ≤ 2 µg/mL according to CLSI. Synergism was observed in 100% of in vitro isolates of CP K. pneumoniae according to Bulman et al. [75]. FOS + polymyxin had a prevalent addictive effect in vitro against Pseudomonas spp. [76] and A. baumannii [65]. In a study there was a complete polymyxin B susceptibility restoration [65]. No antagonistic effect was observed either in in vitro or in vivo studies.
The combination of polymyxins and FOS appears a good option against Enterobacterales and P. aeruginosa strains.

3.11. Daptomycin

Thirteen papers evaluating FOS in combination with daptomycin (DAP) were reviewed (Table 11). DAP is a cyclic lipopeptide administered intravenously for Gram-positive infections, acting through bacterial membrane depolarization [77]. Its breakpoints are ≤1 µg/mL for Staphylococcus spp. and ≤2 µg/mL for Enterococcus spp. [10,78].
When evaluated against S. aureus isolates, the combination FOS + DAP had a synergistic effect in vitro against 37–100% of isolates (synergistic effect of the combination against 100% of the tested isolates was reported in 4 in vitro studies [63,79,80,81] and 2 in vivo studies [37,79]). DAP showed excellent synergistic activity in association with FOS against Enterococcus spp., resulting in synergistic effect in all 34 tested isolates (4 studies). FOS + DAP also exhibited a greater efficacy against E. faecalis biofilm formation than FOS or DAP alone. Efficacy in vivo sometimes differed from the results obtained in vitro, resulting in greater [37] or less [82] efficacy. No antagonistic effect was observed either in in vitro or in vivo studies.
The combination of daptomycin + FOS has good synergistic effect rates against S. aureus and Enterococcus spp. and deserves clinical interest.

3.12. Tigecycline

Fourteen papers evaluating FOS in combination with TIG were reviewed (Table 12). TIG is the first glycylcycline antibiotic, a broad-spectrum class of bacteriostatic derivate from tetracyclines, that acts binding the 30S ribosomal subunits, inhibiting bacterial proteins synthesis. It is only available for intravenous administration and shows activity against either Gram-positive or Gram-negative or atypical bacteria [64]. Its breakpoint are ≤0.5 mg/L both for S. aureus and Enterobacterales and ≤0.25 mg/L for Enterococcus spp. [10].
When evaluated in vitro against Enterobacterales or A. baumannii (10 studies, 338 isolates) FOS + TIG had synergistic effect approximately in 17% of cases and additive effect in the 43%, while indifference was reported for all remaining cases [38,73,74,83,84,85,86,87,88,89]. Furthermore, indifferent effect against all isolates was observed in one in vivo experiment against E. coli [73]. Mostly indifference was observed also when it was tested against N. gonorrhoeae or P. aeruginosa [54,86]. When tested against 61 isolates of Enterococcus spp. (3 studies) many cases of synergistic effect was reported in vitro (about 40% of cases) [55,90,91] and in vivo against E. faecalis [90]. Finally, 2 studies evaluated FOS + TIG combination in vitro against MRSA, but with inconclusive results (total indifference or almost total synergism) [69,90]. In all in vitro studies only 2 cases of antagonism were reported, against K. pneumoniae [89].
According to the literature the combination of TIG + FOS appears to be particularly interesting (good synergistic effect rates) against Enterobacterales and Enterococcus spp.

3.13. Linezolid

Thirteen papers evaluating FOS in combination with linezolid (LZD) were reviewed (Table 13). LZD is a synthetic antibiotic which binds rRNA on both 30S and 50S ribosomal subunits, inhibiting bacterial proteins synthesis [92]. It is used for Gram-positive infections treatment, including MRSA and E. faecium vancomycin-resistant (VREF) infections [93]. Its breakpoint is ≤4 µg/mL both for S. aureus and E. faecium.
When evaluated against S. aureus isolates (9 studies), combination FOS + LZD had a synergistic effect in vitro approximately in 95% of cases (synergistic effect of the combination against 100% of the tested isolates was reported in 6 in vitro studies [36,43,63,94,95]) and even against staphylococcal biofilm cultures [69]; furthermore, the only 2 in vivo studies performed proved FOS + LZD combination to have higher efficacy than FOS or LZD alone [36,95]. One study evaluated the combination on 2 strains of S. epidermidis proving synergism on both [43]. Otherwise, in the 4 studies in which it was tested against E. faecium, this combination showed in most cases additive effect and only few cases of synergism. In no case was reported synergistic effect against E. faecalis (2 studies). No antagonistic effect was observed either in in vitro or in vivo studies.
The good synergistic effects reported make LZD + FOS a promising combination against staphylococci.

3.14. Rifampin

Fourteen papers evaluating FOS in combinations with rifampin were reviewed (Table 14). Rifampin breakpoints are ≤0.06 µg/mL for Staphylococcus spp., Streptococcus spp. and ≤0.125 µg/mL for S. pneumoniae. Rifampin inhibits bacterial DNA-dependent RNA polymerase with a concentration related effect. It is used for the treatment of intracellular pathogens and it has a broad-spectrum antibacterial activity. Rifampin breakpoints are not defined either by EUCAST or by CLSI for Acinetobacter spp., Enterobacterales and Enterococcus spp. Based on literature data, susceptibility was defined as a MIC ≤ 1 µg/mL for Enterococcus spp. [71]. Rifampin showed synergistic activity in association with FOS against Enterococcus spp., resulting in synergistic effect in 20−100% of cases. High activity was reported in vitro and in vivo in a recent paper where FOS + RIFA also exhibited a greater efficacy against E. faecalis biofilm formation [90]. When evaluated against S. aureus isolates, the combination FOS + rifampin had a synergistic effect in vitro against 34−100% of isolates. Synergistic effect of the combination against 100% of the tested isolates was reported in 3 in vitro studies [43,90,96] and 2 in vivo studies [37,96]. Antagonistic effect was observed only in 33% of isolates in the study by Quentin et al. [35] where the antibiotic combination was antagonist for the isolates susceptible and intermediate to rifampin and indifferent for those resistant. No antagonistic effect was observed in other studies.
In clinics RIF + FOS should be considered (usually with a third agent) against S. aureus sustained infections, especially when biofilm production is likely.

3.15. Miscellanea

Two papers evaluating FOS in combination with metronidazole (MTZ) were reviewed (Table S1). MTZ is a bacteriostatic antimicrobial, active on bacteria (mainly anaerobic) and parasites. When evaluated in vitro against Helicobacter pylori, combination FOS + MTZ had a prevalent indifferent effect, an additive effect in only 21% of cases and an antagonist effect in 4% [97]. In vivo study showed a significantly decrease mortality and increase cure rates if the animal treated with MTZ + FOS [98].
One paper evaluating FOS in combination with spectinomycin (SCM) was reviewed (Table S1). SCM is an aminocyclitol aminoglycoside antibiotic with bacteriostatic activity, used to treat gonorrhea. In vitro study reported that antimicrobial combinations of SMC + FOS no synergistic effect was found [54].
One paper evaluating FOS in combination with sulbactam (SLB) was reviewed (Table S1). SLB is an irreversible β-lactamase inhibitor capable to binding to penicillin-binding proteins and with weak antimicrobial activity. When evaluated in vitro against A. baumannii OXA-23, combination FOS + SLB had a synergistic effect in 75% of case, and an indifferent effect in 25% of cases [99].
One paper evaluating FOS in combination with lincomycin (LNM) was reviewed (Table S1). LMN is a protein synthesis inhibitor with activity against gram positive and anaerobic bacteria. When evaluated in vitro against S. aureus, combination FOS + LNM had a synergistic effect in 81% of case and an additive effect in 25% of cases [14].
One paper evaluating FOS in combination with nitroxoline (NTX) was reviewed (Table S1). NTX is a urinary antibacterial agent active against susceptible Gram-positive and Gram-negative organisms. In vitro study, NTX was synergistic with FOS in only 12% of cases and in other cases shoed an indifferent effect (88%) [66].
Two papers evaluating FOS in combination with quinupristin/dalfopristin (Synercid) were reviewed (Table S1). Synercid is a protein synthesis inhibitor used to treat infections by staphylococci and by vancomycin-resistant strain. When evaluated in vitro against methicillin resistant or susceptible Staphyloccoccus spp., combination FOS + Synercid had a synergistic effect in 100% of case [43,100].
Three papers evaluating FOS in combination with fusidic acid (FSA) were reviewed (Table S1). FSA is a bacteriostatic antibiotic with acts as a bacterial protein synthesis inhibitor. When evaluated in vitro against MRSA, combination FOS + FSA had a various behavior, showing a synergistic effect in 88–100% of case or an indifferent effect in 100% of cases. No antagonism was found [69,101,102].
Four papers evaluating FOS in combination with chloramphenicol (CHL) were reviewed (Table S1). CHL is a synthetic broad-spectrum antimicrobial, mainly bacteriostatic, active on numerous Gram-positive and Gram-negative, aerobic and anaerobic bacteria; it acts binding 50S ribosomal subunit, inhibiting bacterial protein synthesis [103]. Its breakpoint is ≤ 8 mg/L both for S. aureus and Enterobacterales [10]. When evaluated in vitro against either Enterobacterales (468 isolates, 4 studies), combination FOS + CHL had synergistic effect approximately in 40% of cases, while additive effect in 35% and indifferent effect in the remaining cases [14,66,104,105]. Furthermore, one study tested this combination against S. aureus, with similar results (synergistic effect against 44% of isolates) [14]. No antagonistic effect was observed.
Three papers evaluating FOS in combination with trimethoprim-sulfamethoxazole (TMP-SMX) were reviewed (Table S1). TMP-SMX is a fixed combination of 2 antimicrobials that inhibits bacterial synthesis of tetrahydrofolate, a necessary cofactor for bacterial DNA synthesis. It is available in oral or intravenous preparation and it is mainly used for treatment of urinary and respiratory infections [106]. Its breakpoint is ≤ 2 µg/mL both S. aureus and Enterobacterales [10]. When evaluated in vitro against either S. aureus (148 isolates) or Enterobacterales (120 isolates), combination FOS + TMP-SMX had indifferent effect approximately against 92% of isolates [12,38,66]. Only in few cases, against Enterobacterales, was reported synergistic or additive effect (1 study) [38] and even antagonistic effect was reported in 4 cases when tested against S. aureus [12].
Two papers evaluating FOS in combination with nitrofurantoin (NTF) were reviewed (Table S1). NTF is a synthetic antibiotic administered orally mainly for treatment of lower urinary tract infections. Its breakpoint is ≤ 64 µg/mL both E. faecalis and Enterobacterales [10]. When evaluated in vitro against either vancomycin-resistant E. faecium (32 isolates) or Enterobacterales (100 isolates), combination FOS + NTF had indifferent effect against 100% of isolates [66,67]. No synergistic, additive or antagonistic effect was observed.

3.16. Non-Antibiotic Molecules

One paper evaluating FOS in combination with auranofin (AF) was reviewed (Table S2). AF is an orally active gold compound for the treatment of rheumatoid arthritis. When evaluated in vitro against Staphyloccoccus spp., combination FOS + AF had showed a reduction of bacterial load for both MSSA and MRSA strains. In vivo, this combination had showed a synergistically inhibition of abscess and inflammation formation. No interactions were showed against S. epidermidis MS [107]. Three paper evaluating FOS in combination with dilipid ultrashort cationic lipopeptides, tobramycin-efflux pump inhibitor (TOB-EPI) conjugates or amphiphilic lysine-tobramycin conjugates (ALT) against P. aeruginosa, were reviewed (Table S2). For all combinations, in vitro studies had showed a synergistic effect (100%). Furthermore, in presence of TOB-EPI or ALT conjugates MICs of FOS were dramatically reduced [108,109,110]. One paper evaluating FOS in combination with β-chloro-L-alanine (β-CLA) was reviewed (Table S2). β-CLA is an amino acid analog of FOS. When evaluated in vitro against MRSA, combination FOS + β-CLA had showed a synergistic effect on biofilm production [111]. One paper evaluating FOS in combination with plectasin NZ2114, compound capable to inhibits a cell wall biosynthesis, was reviewed (Table S2). When plectasin NZ2114 evaluated in vitro against E. faecalis, in combination with FOS it no show a synergistic effect [112]. One paper evaluating FOS in combination with 2 quinolone derivatives (A and B) was reviewed (Table S2). When evaluated in vitro against E. faecalis VRE and MRSA, combination FOS + A had always showed a synergistic effect, while FOS + B had showed a synergistic effect in 64% of cases and in other cases shoed an additive effect (36%) [113]. One paper evaluating FOS in combination with N-acetylcysteine (NAC), a mucolytic agent, was reviewed (Table S2). The in vitro analysis against E. coli, had showed a capable of NAC to reduce biofilm if used in combination with FOS. The most effective combination was that obtained using FOS at 2000 mg/L and NAC at 2 mg/mL [114]. One paper evaluating FOS in combination with sophoraflavanone G (SFG), a phytoalexins, was reviewed (Table S2). When evaluated in vitro against MRSA, combination FOS + SFG had showed a synergistic effect (100%) [115]. One paper evaluating FOS in combination with arenaemycin (ARM), also called pentalenolactones, was reviewed (Table S2). When evaluated in vitro against P. vulgaris and S. gallinarum, combination FOS + ARM had showed a synergistic effect (100%) [116]. One paper evaluating FOS in combination with chlorogenic acid (CHA) and caffeic acid (CFA) was reviewed (Table S2). When evaluated in vitro against Resistant Listeria monocytogenes, combination FOS + CHA had showed a reduction in the cell growth equal to 98% and FOS + CFA as to 85,2%. Moreover, CHA restored a FOS susceptibility in 100%, if 3 mg/L [117]. One paper evaluating FOS in combination with silver (AgNPs) and zinc oxide (ZnONPs) nanoparticles, are molecules known to affect bacterial membranes, was reviewed (Table S2). When evaluated in vitro against S. aureus, S. enterica, and E. coli, combination FOS + AgNPs or ZnONPs had showed a synergistic effect (100%) [118].

4. Discussion

FOS is an inhibitor of bacterial wall synthesis with a unique mechanism of action. Its use in clinic is increasing as is often active against MDR bacteria. Intravenous FOS is often administered in combination with other antibiotics therefore the knowledge of pharmacodynamic interactions is of fundamental importance. In this review, we have investigated the role of FOS as partner drug, by analyzing literature studies in which it has been used in vitro and in vivo in combination with other antibiotics and evaluating the antimicrobial activity of combinations against the most common bacterial pathogens. From this huge data collection, no clinically significant antagonistic effect came out between FOS and any most common used antibiotics for the treatment of nosocomial infections.
FOS has been studied in combination with the major antibiotic classes (penicillins, cephalosporins, carbapenems, monobactams, quinolones, aminoglycosides, macrolides, glycopeptides, tetracyclines, polimyxins, lipopeptides, oxazolydinones, and rifampicin) against both Gram-negative and Gram-positive bacteria. A total of 185 literature reports accounted for 9,927 study isolates. FOS-based synergistic interactions were detected in 33.7% of total isolates, although additive and indifferent interactions were more prevalent (65.4%). Antagonism occurred sporadically (0.9% of total isolates).
Clinically significant synergistic interactions were mostly distributed in combination with penicillins (51%), carbapenems (43%), chloramphenicol (39%), and cephalosporins (33%) in Enterobactaerales; with linezolid (74%), tetracyclines (72%), and daptomycin (56%) in S. aureus; with chloramphenicol (53%), aminoglycosides (43%) and cephalosporins (36%) against P. aeruginosa; with daptomycin (97%) in Enterococcus spp. and with sulbactam (75%) and penicillins (60%) and in Acinetobacter spp.
Notably, 31.2% of synergistic interactions occurred in Enterobacterales (FOS in combination with 3 different antibiotics), followed by 31% occurred in S. aureus (FOS in combination with 4 different antibiotics) and 7.6% occurred Enterococcus spp. (FOS in combination with 5 different antibiotics).
From a clinical point of view, taking into account the antimicrobial stewardship principles and the priorities in terms of MDR impact, our work points out good pharmacodynamic interactions rates (additive/synergistic effects) when FOS is especially combined with:
(1)
Cephalosporins and cephalosporins + β-lactamase inhibitors, including ceftazidime/avibactam and ceftolozane/tazobactam, for Enterobacterales and P. aeruginosa;
(2)
carbapenems for K. pneumoniae and P. aeruginosa;
(3)
quinolones for P. aeruginosa;
(4)
polymyxins for K. pneumoniae;
(5)
daptomycin for Staphylococcus spp (MRSA included), and Enterococcus spp.;
(6)
linezolid for Staphylococcus spp.; and
(7)
sulbactam for A. baumannii.
When FOS is combined with molecules other than antibiotics, chlorogenic acid and caffeic acid appeared to be good partner drugs against L. monocytogenes.
Our tables (including the summarizing Table 15) could act as a useful consultation tool for clinicians using FOS both as empirical or targeted antibiotic regimen.

5. Conclusions

In conclusion, taken together, these data, the pharmacological characteristics (i.e., excellent distribution in body sites, the safety and tolerability profile) and the encouraging positive clinical outcome of treated patients highlight the role of FOS as partner drug (mostly intravenously) for the treatment of infections caused by common (including MDR) pathogens. In particular, the presence of synergistic interactions and the almost total absence of antagonisms, make FOS a good partner drug in clinical practice. Moreover, improving FOS-based combinations could act as a meropenem- and colistin-sparing agent, mostly contributing to prevent AMR, especially related to last resource antibiotics.
Table 1. Studies on combination between fosfomycin and penicillins, penicillins + β-lactamase inhibitors, penicillinase-resistant penicillins. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 1. Studies on combination between fosfomycin and penicillins, penicillins + β-lactamase inhibitors, penicillinase-resistant penicillins. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorPenicillinNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)Fosfomycin-Resistant (%)penicillin-Resistant (%)In Vitro (methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)Fosfomycin Susceptibility Restoration (%)Penicillin Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAAverypiperacillin/tazobactam498 E. coli: KPC (25%), NDM (75%), ESBL (62.5%); 35 Klebsiella spp: KPC (45.7%), NDM (40%); OXA (14.3%), VIM (8.6%), ESBL (88.6%), fosA (44%); 2 Citrobacter spp: KPC (50%), NDM (50%), ESBL (50%), 4 E. cloacae: KPC (75%), NDM (25%), ESBL (75%)20 (40.8%)49 (100%)in vitro (ET)1 (2%)2 (4%)46 (94%)0%--Data on synergism reported without distinction for bacterial strains. % of FOS-R isolates estimated on the basis of the reported MIC50.[11]
2019, USAFlammpiperacillin/tazobactam20---in vitro (CB, TK)12 (60%)7 (35%)0%0%--For 1 isolate the efficacy of FOS + PIP/TAZ remained indeterminate.[38]
1978, SpainOlayampicillin, carbenicillinAmpicillin: 17 E. coli, 11 Klebsiella spp., 7 E. cloacae, 14 Proteus spp., 22 Salmonella spp. Carbenicillin: 16 E. coli, 32 S. marcescens, 26 Proteus spp.---in vitro (CB)ampicillin: 31 (43%); carbenicillin: 24 (32%)ampicillin: 31 (43%); carbenicillin: 31 (41%)ampicillin: 9 (12%); carbenicillin: 19 (25%)0%---[14]
E. coli2020, KoreaSeokpiperacillin/tazobactam2ESBL (100%)0%1 (50%)in vitro (TK)0%0%2 (100%)0%---[119]
2018, FranceBerleurtemocillin3KPC (33.3%), OXA (33.3%)0%Breakpoints NAin vitro (CB, TK); in vivo (mouse, peritonitis)0%in vitro: 3 (100%); in vivo: 3 (100%)0%0%---[15]
2014, SwedenHickammecillinam2ESBL, OXA (50%)0%0%in vitro (CB, TK)2 (100%)0%0%0%---[120]
1977, PolandBorowskiampicillin10---in vitro (CB)7 (70%)1 (10%)2 (20%)0%---[121]
K. pneumoniae2014, SwedenHickammecillinam1ESBL, OXA (100%)0%0%in vitro (CB, TK)1 (100%)0%0%0%---[120]
Salmonella spp.1977, SpainPereaampicillin90-17 (18.9%)11 (12%)in vitro (CB, TK)74 (82%)7 (7%)7 (7%)0%--For 2 isolates the effect of FOS + ampicillin remained indeterminate. The authors considered synergistic the effect for FICI up to 0.75.[104]
1977, SpainFigueroaampicillin16---in vitro (CB)15 (93%)1 (6%)0%0%--S. typhi. The authors considered synergistic the effect for FICI up to < 1. They also evaluated different antibiotic combinations on patients with typhoid fever: FOS + AMP resulted in the highest rate of cures.[105]
Shigella spp.1977, SpainPereaampicillin50-27 (54%)30 (60%)in vitro (CB, TK)27 (54%)9 (18%)14 (28%)0%--The authors considered synergistic the effect for FICI up to 0.75.[104]
P. aeruginosa2019, USAAverypiperacillin/tazobactam103-NA (at least 71)103 (100%)in vitro (ET)3 (2%)26 (25%)74 (71%)0%-15 (14.6%)-[33]
2019, USAFlammpiperacillin/tazobactam5---in vitro (CB, TK)0%5 (100%)0%0%---[38]
2013, Brazildos Santospiperacillin/tazobactam4-4 (100%)2 (50%)in vitro (CB)4 (100%)0%0%0%2 (50%)1 (50%)-[48]
2002, JapanOkazakipiperacillin30-15 (50%)30 (100%)in vitro (efficacy time index)3 (10%)6 (20%)21 (70%)0%0%15 (50%)-[39]
1984, JapanTakahashipiperacillin20---in vitro (CB)4 (20%)16 (80%)0%0%---[122]
1978, SpainOlaycarbenicillinin vitro: 73; in vivo: 2---in vitro (CB); in vivo (mouse, peritonitis)in vitro: 21 (28%); in vivo: 2 (100%)in vitro: 40 (54%)in vitro: 12 (16%)0%---[14]
Acinetobacter spp.2019, USAFlammpiperacillin/tazobactam5 (A. baumannii-calcoaceticus species complex)---in vitro (CB, TK)3 (60%)1 (20%)0%0%--For 1 isolate the efficacy of FOS + PIP/TAZ remained indeterminate.[38]
S. aureus2015, Spaindel Ríoamoxicillin + clavulanic acid10Methicillin-resistant Staphylococcus aureus (MRSA) (100%)1 (10%)10 (100%)in vitro (TK)in vitro: 8 (80%); in vivo: 2 (100%)in vitro: 2 (20%)0%0%---[28]
2003, JapanNakazawaampicillin32MRSA (100%)29 (91%)31 (96%)in vitro (efficacy time index)4 (12%)2 (6%)26 (81%)0%---[18]
1997, ItalyFerraraoxacillin16MRSA (100%)NA (at least 8)16 (100%)in vitro (TK)3 (18%)3 (18%)4 (25%)---Addition or indifference was observed for the remaining 6 strains (data not shown).[123]
1994, JapanKomatsuzawaoxacillin38MRSA (60.5%)33 (86.8%)23 (60%)in vitro (CB)20 (52%)17 (44%)1 (2%)0%---[124]
1985, USAAlvarezmethicillin148MRSA (100%)NA (< 15)148 (100%)in vitro (CB)69 (46%)--1 (1%)--For the 78 remaining strains it was not specified if the combination FOS + methicillin acted with an additive or indifferent effect.[12]
1978, SpainOlayampicillin, carbenicillinampicillin: 27; carbenicillin: 28---in vitro (CB)ampicillin: 15 (55%); carbenicillin: 10 (35.7%)ampicillin: 9 (33%); carbenicillin: 18 (64%)ampicillin: 3 (11%); carbenicillin: 0%0%---[14]
1977, PolandBorowskipenicillin G11---in vitro (CB)5 (45%)2 (18%)4 (36%)0%---[121]
S. epidermidis1997, ItalyFerraraoxacillin12MRSE (100%)NA (at least 6)12 (100%)in vitro (TK)6 (50%)1 (8%)1 (8%)---Data of the other 4 strains are not shown.[123]
Streptococcus spp.2017, GermanyGonzalez Morenobenzylpenicillin3-1 (33.3%)0%in vitro (microcalorimetry for biofilms)0%0%3 (100%)0%--S. agalactiae, S. pyogenes, S. oralis. High-dose FOS caused a delay of 8 h in the production of heat, compared with untreated controls, suggesting that the treatment could result in a reduction in the number of viable sessile cells, although not in complete biofilm eradication.[9]
1981, SpainVicentepenicillin G17-9 (53%)5 (29%)in vitro (CB, TK); in vivo (rabbit, endocarditis)in vitro: 4 (23%)in vitro: 12 (71%); in vivo: 100%in vitro: 1 (6%)0%--S. sanguis. The mean log10 CFU per gram of vegetations in the FOS + penicillin groups was significantly lower than that in the FOS groups but was not significantly lower than that in the penicillin group.[17]
1978, SpainOlayampicillin37---in vitro (CB)12 (32%)11 (29%)14 (37%)0%---[14]
S. pneumoniae2001, SpainBañón Ariaspenicillin10-1 (10%)8 (80%)in vitro (TK)10 (100%)0%0%0%--Synergistic effect difficult to determine. It is reported as synergistic against all isolates based on authors’ considerations and on the comparison between cumulative efficacy of MIC + MIC and MIC/4 + MIC/4.[125]
1996, FranceChavanetamoxicillin1-0%1 (100%)in vivo (rabbit, fibrin clot infection)1 (100%)0%0%0%---[23]
1995, JapanKikuchibenzylpenicillin51-0%51 (100%)in vitro (CB, TK)9 (17%)42 (82%)0%0%---[126]
Enterococcus spp.2013, TaiwanTangampicillin10 E. faecium, 9 E. faecalisVRE (100%)13 (68%)9 (47%)in vitro (TK, biofilm)TK: 3 (15%)--biofilm: 6 (31%)--The 3 isolates exhibiting synergistic effect were all E. faecium. The 6 isolates exhibiting antagonistic effect on biofilm formation were all E. faecalis. From the data reported in the paper it was not possible to establish the effect of the combination against the other isolates.[13]
1995, FrancePestelpenicillin10-10 (100%)6 (60%)in vitro (CB, TK)6 (60%)--0%--E. faecalis, E. faecium, E. casseliflavus, E. durans. The authors did not distinguish between additive and indifferent effect.[127]
E. faecalis2011, ItalyFarinaampicillin27-2 (7%)0%in vitro (ET)2 (7%)0%25 (92%)0%--The Authors considered 0.5 < FICI ≤ 4 as indifferent.[128]
E. faecium2013, USADescourouezamoxicillin4VRE (100%)0%4 (100%)in vitro (TK)100%0%0%0%--The combination resulted also strongly bactericidal.[67]
Table 2. Studies on combination between fosfomycin and cephalosporins, cephalosporins + β-lactamase inhibitors. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 2. Studies on combination between fosfomycin and cephalosporins, cephalosporins + β-lactamase inhibitors. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorCephalosporinNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)cephalosporin-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Cephalosporin Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAAverycefepime (FEP), ceftolozane/tazobactam (C/T), ceftazidime (CTZ), ceftazidime/avibactam (CZA)49 (26 tested for CZA)8 E. coli: KPC (25%), NDM (75%), ESBL (62%); 35 Klebsiella spp: KPC (45%), NDM (40%); OXA (14%), VIM (8%), ESBL (88%), fosA (44%); 2 Citrobacter spp: KPC (50%), NDM (50%), ESBL (50%), 4 E. cloacae: KPC (75%), NDM (25%), ESBL (75%)20 (40%)49 (100%)in vitro (ET)FEP: 2 (4%); C/T: 8 (16%); CTZ: 3 (6%); CZA: 0%FEP: 5 (10%); C/T: 11 (22%); CTZ: 8 (16.3%); CZA: 3 (11.5%)FEP: 42 (85%); C/T: 30 (61%); CTZ: 38 (77%); CZA: 23 (88%)0%0%0%Data on synergism reported without distinction for bacterial strains. % of FOS-R isolates estimated on the basis of the reported MIC50.[11]
2019, USAFlammceftazidime20---in vitro (CB, TK)8 (40%)10 (50%)0%0%--For 2 isolates the efficacy of FOS + CTZ remained indeterminate.[38]
1978, SpainOlaycephalexin23 E. coli, 29 Salmonella spp., 8 Klebsiella spp., 11 E. cloacae, 16 S. marcescens, 16 Proteus spp.---in vitro (CB)42 (40%)46 (44%)15 (14%)0%---[14]
E. coli2020, KoreaSeokcefixime4ESBL (50%)0%2 (50%)in vitro (TK)4 (100%)0%0%0%---[119]
2014, FranceLefortcefoxitim2ESBL (50%)0%breakpoints NAin vitro (TK); in vivo (mouse, urinary tract infection)in vitro: 2 (100%); in vivo: 2 (100%)0%0%0%---[30]
K. pneumoniae2019, PolandOjdanaceftazidime-avibactam19NDM (52%); KPC (42%); OXA (5%)10 (53%)10 (53%)in vitro (ET)9 (47%)7 (36%)3 (15%)0%---[31]
2019, USAMikhailceftazidime-avibactam21fosA/fosA-like, KPC, ESBL, OXA (100%)15 (71%)0%in vitro (CB, TK)10 (47%)9 (42%)2 (9%)0%-0% (all S)It is reported only the reduction of CZA in combination and time–kill was performed only on 2 isolates randomly selected, therefore a reduction of at least 4 times was considered as synergistic. A 2-fold reduction was considered as additive. No reduction was considered as indifferent. In increase of MIC in combination was considered antagonistic.[21]
1977, SpainDazacephapirin33-100%breakpoints NAin vitro (CB)1 (3%)---0%Breakpoints NA (reduction of MIC from 16 to 4 µg/mL)The authors reported only the number of isolates on which the combination had a synergistic effect.[66]
P. aeruginosa2020, BrazilCubaceftolozane/tazobactam27carbapenemase-producing (74%)26 (96%)22 (81%)in vitro (ET, TK)24 (88%)3 (11%)0%0%24 (92%)-It is not possible to establish the % of strains with FOS susceptibility restoration because the MIC for all R strains was > 64 ug/mL and it is not reported the MIC in combination but the MIC fold reduction. It is however strongly reduced (range: 2–16 fold reduction).[32]
2020, USAMullanecefepime, ceftolozane/tazobactam28 CEF; 15 C/T---in vitro (CB, TK)CEF: 5 (18%); C/T: 5 (33%)CEF: 20 (71%); C/T: 8 (53%)CEF: 3 (11%); C/T: 2 (14%)0%-CEF: 1 (4%); C/T: 5 (33%)-[129]
2019, USAMikhailceftazidime-avibactam21fosA/fosA-like, KPC, ESBL, OXA (100% at least 1 resistance gene)19 (90%)5 (23%)in vitro (CB, TK)7 (33%)6 (28%)8 (38%)0%-1 (20%)It is reported only the reduction of CZA in combination and time–kill was performed only on 2 isolates randomly selected, therefore a reduction of at least 4 times was considered as synergistic. A 2-fold reduction was considered as additive. No reduction was considered as indifferent. In increase of MIC in combination was considered antagonistic.[21]
2019, USAPapp-Wallaceceftazidime-avibactam1-0%1 (100%)in vitro (CB, TK); in vivo (mouse)in vitro: 100%; in vivo: 100%0%0%0%---[29]
2019, USAAverycefepime (FEP), ceftolozane/tazobactam (C/T), ceftazidime (CAZ), ceftazidime/avibactam (CZA)92 FEP, 14 C/T, 81 CAZ, 16 CZACarbapenem-resistant (100%)-100%in vitro (ET)FEP: 22 (23%); C/T: 7 (50%); CAZ: 42 (51%); CZA: 4 (25%)FEP: 53 (57%); C/T: 5 (35%); CAZ: 31 (38%); CZA: 12 (75%)FEP: 17 (18%); C/T: 2 (14%); CAZ: 8 (9%); CZA: 0%0%-FEP: 56 (60%); C/T: 10 (71%); CAZ: 46 (56%); CZA: 11 (68%)-[33]
2019, USAFlammceftazidime5---in vitro (CB, TK)2 (40%)3 (60%)0%0%---[38]
2018, USAMonogueceftolozane/tazobactam4-3 (75%)2 (50%)in vitro (TK)1 (25%)2 (50%)1 (25%)0%---[34]
2013, Brazildos Santosceftazidime3-3 (100%)3 (100%)in vitro (CB)3 (100%)0%0%0%1 (33%)2 (66%)-[48]
2005, ThailandPruekprasertceftazidime18---in vitro (CB)2 (11%)6 (33%)6 (33%)4 (22%)---[22]
2002, JapanOkazakiceftazidime, cefepime30-15 (50%)CAZ: 28 (93%), CEFP: 26 (86.7%)in vitro (efficacy time index)CAZ: 21 (70%); CEFP: 24 (80%)CAZ: 8 (26%); CEFP: 1 (3.3%)CAZ: 1 (3%); CEFP: 5 (16%)0%CAZ: 3 (20%); CEFP: 6 (40%)CAZ: 19 (67%); CEFP: 26 (100%)-[39]
1999, JapanHayamiceftazidime26-NA (at least 13)NA (at least 5)in vitro (CB, TK)7 (26%)14 (53%)5 (19%)0%---[130]
1997, FranceTessierceftazidime40-21 (52%)14 (35%)in vitro (CB)0%8 (20%)32 (80%)0%20 (95%)8 (57%)Although the combination had a synergistic effect on no tested strains, it is of clinical relevance as it restored FOS and CTZ susceptibility in many resistant isolates.[131]
1984, JapanTakahashicefoperazone, cefsulodin20 (cefoperazone), 23 (cefsulodin)---in vitro (CB)cefoper: 17 (85%); cefsul: 19 (92%)cefoper: 3 (15%); cefsul: 4 (17%)0%0%---[122]
A. baumannii2019, USAFlammceftazidime5 (A. baumannii-calcoaceticus species complex)---in vitro (CB, TK)2 (40%)1 (20%)1 (20%)0%--For 1 isolate the efficacy of FOS + CTZ remained indeterminate.[38]
1996, SpainMartinez-Martinezceftazidime34-34 (100%)32 (94%)in vitro (CB)1 (3%)NANA0%--Only synergistic and antagonistic effect reported.[132]
Staphylococcus spp.1995, ItalyMarchesecefdinir6 S. aureus, 8 S. epidermidis, 2 S. hominis, 2 S. xylosus, 5 S. saprophyticus, 2 S. haemolyticusPenicillin-resistant (100%)--in vitro (CB, TK)4 (16%)--0%--The authors considered 0.5 < FICI ≤ 4 as indifferent, therefore it is not possible to establish if the effect was additive or indifferent for most strains.[114]
S. aureus2003, JapanNakazawaflomoxef sodium (FS), cefmetazole (CEM), cefotiam (CET), cefoperazone/sulbactam (CS)32MRSA (100%)29 (91%)FS: 29 (91%); CEM: 16 (50%); CET: 30 (94%); CS: 27 (84%)in vitro (efficacy time index)FS: 7 (22%); CEM: 26 (81%); CET: 7 (22%); CS: 19 (59%)FS: 11 (34%); CEM: 3 (9%); CET: 1 (3%); CS: 8 (25%)FS: 14 (44%); CEM: 3 (9%); CET: 22 (69%); CS: 5 (15%)0%---[18]
1978, SpainOlaycephalexin24---in vitro (CB)17 (70.8%)7 (29.2%)0%0%---[14]
2015, Spaindel Ríoceftriaxonein vitro 10; in vivo 2MRSA (100%)1 (10%)10 (100%)in vitro (TK); in vivo (rabbit, endocarditis)in vitro: 8 (80%); in vivo: 2 (100%)in vitro: 2 (20%)0%0%--% of sterile vegetations: FOS alone 0%, IMI alone 0%, FOS + CRO 62%.[28]
1985, GermanyPortiercefotaxime, cephalotin, cefoperazone, cefamandole10MRSA (100%)0%10 (100%)in vitro (CB)cefotaxime, cephalotin, cefoperazone, cefamandole: 10 (100%)0%0%0%---[20]
1990, FranceChavanetcefotaxime1MGRSA (100%)0%1 (100%)in vivo (rabbit, subcutaneous fibrin clots)1 (100%)0%0%0%--Synergistic effect was observed when both drugs were administered in two divided doses.[27]
1985, FranceKazmierczakcefotaxime1-0%1 (100%)in vivo (rabbit, meningitis)0%1 (100%)0%0%--Cefotaxime: variable drop in bacterial numbers from one rabbit to another during the first 12 h, then a bacteriostasis. FOS: rapid bactericidal effect during the first 12 h, becoming slower during the following 36 h (0.03% surviving bacteria at 48 h). Cefotaxime + FOS: rapid bactericidal effect remaining steady over the 48-h period (0.001% surviving bacteria at 48 h).[26]
1991, JapanMatsudacefmetazole25MRSA (100%)25 (100%)25 (100%)in vitro (CB, TK)11 (44%)11 (44%)3 (12%)0%---[133]
1986, JapanUtsuicefmetazole14 in vitro, 7 in vivoMRSA (100%)-14 (100%)in vitro (CB, TK); in vivo (mouse)in vitro: 10 (71%); in vivo: 5 (71%)in vitro: 4 (28%); in vivo: 2 (28%)0%0%---[25]
1987, FranceCourcolceftriaxone6-1 (16.%)6 (100%)in vitro (CB, TK)CB: 1 (16%); TK: 1 (16%)CB: 0%; TK: -CB: 4 (66%); TK: 3 (50%)CB: 1 (16%); TK: ---Different activity of the drug combination with checkerboard assay or time–kill assay. The effect of FOS + ceftriaxone on 2 isolates remained indeterminate. The authors considered the combination antagonistic when the FICI was > 2.[19]
1985, USAAlvarezcefamandole148MRSA (100%)NA (<15)-in vitro (CB)97 (66%)--0%--For the 78 remaining isolates it was not specified if the combination FOS + cefamandole acted with an additive or indifferent effect.[12]
2001, AustriaGrifcefazolin5MRSA (20%), GISA (20%)--in vitro (CB, TK)5 (100%)0%0%0%---[43]
S. epidermidis2001, AustriaGrifcefazolin2---in vitro (CB, TK)0%0%2 (100%)0%---[43]
1987, FranceCourcolceftriaxone6-2 (33.3%)6 (100%)in vitro (CB, TK)CB: 1 (16%); TK: 5 (83.3%)CB: 0%; TK: -CB: 5 (83%); TK: -CB: 1 (16%); TK: ---Different activity of the drug combination with checkerboard assay or time–kill assay. The effect of FOS + ceftriaxone on 1 isolate remained indeterminate. The authors considered the combination antagonistic when the FICI was > 2.[19]
S. pneumoniae2006, SpainRibesceftriaxone2-0%2 (100%)in vitro (TK); in vivo (rabbit, meningitis)0%in vitro: 1 (50%); in vivo: 2 (100%)in vitro: 1 (50%)0%---[24]
2001, SpainBañón Ariasceftriaxone10-1 (10%)7 (70%)in vitro (TK)10 (100%)0%0%0%--Synergistic effect difficult to determine. It is reported as synergistic against all isolates based on authors’ considerations and on the comparison between cumulative efficacy of MIC + MIC and MIC/4 + MIC/4.[125]
1994, FranceDoitceftriaxone26-0%20 (76%)in vitro (TK)0%26 (100%)0%0%---[134]
1993, FranceBarakettcefotaxime7-0%2 (28%)in vitro (TK)3 (42%)1 (14%)3 (42%)0%---[135]
1995, FranceChavanetcefotaxime, ceftriaxone1-0%1 (100%)in vitro (TK); in vivo (rabbit, fibrin clot infection)in vitro: 0%; in vivo: 1 (100%, cefotaxime)in vitro: 1 (100%, both cefotaxime and ceftriaxone); in vivo: 1 (100%, ceftriaxone)0%0%---[23]
S. sanguis1981, SpainVicentecefoxitim17-9 (53%)3 (16%)in vitro (CB, TK); in vivo (rabbit, endocarditis)in vitro: 8 (47%); in vivo: 100%in vitro: 8 (47%)in vitro: 1 (6%)0%--The mean log10 CFU per gram of vegetations in the FOS + cefoxitim groups was significantly lower than that in the FOS groups and in the cefoxitim groups.[17]
Enterococcus spp.1995, FrancePestelcefotaxime50-48 (96%)50 (100%)in vitro (CB, TK)45 (90%)-5 (10%)0%--E. faecalis, E. faecium, E. casseliflavus, E. durans. The authors did not distinguish between additive and indifferent effect.[127]
E. faecalis2011, ItalyFarinaceftriaxone27-2 (7%)27 (100%)in vitro (ET)15 (55%)0%12 (44%)0%--The authors did not distinguish between additive and indifferent effect, considering 0.5 < FICI ≤ 4 as indifferent.[128]
N. gonorrhoeae2015, SwitzerlandHauserceftriaxone8-0%1 (12.5%)in vitro (CB)0%0%8 (100%)0%---[57]
2015, The NetherlandsWindcefixime, ceftriaxone4---in vitro (ET)0%cefixime: 1 (25%); ceftriaxone: 2 (50%)cefixime: 3 (75%); ceftriaxone: 2 (50%)0%---[54]
2014, USABarbeecefixime, ceftriaxone32-0%cefotaxime: 29 (90%), cefixime: 6 (18%), ceftriaxone: 0%in vitro (ET)0%0%32 (100%)0%--The authors did not distinguish between additive and indifferent effect, considering 0.5 < FICI ≤ 4 as indifferent.[136]
Table 3. Studies on combination between fosfomycin and carbapenems. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 3. Studies on combination between fosfomycin and carbapenems. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainsYear and CountryAuthorCarbapenemNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Carbapenem-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%) Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Carbapenem Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAAverymeropenem498 E. coli: KPC (25%), NDM (75%), ESBL (62%); 35 Klebsiella spp: KPC (45%), NDM (40%); OXA (14%), VIM (8%), ESBL (88%), fosA (44%); 2 Citrobacter spp: KPC (50%), NDM (50%), ESBL (50%), 4 E. cloacae: KPC (75%), NDM (25%), ESBL (75%)20 (40.8%)49 (100%)in vitro (ET)1 (2%)10 (20%)38 (77%)0%--Data on synergism reported without distinction for bacterial strains. % of FOS-R isolates estimated on the basis of the reported MIC50.[11]
2019, USAFlammmeropenem20---in vitro (CB, TK)8 (40%)10 (50%)0%0%--For 2 isolates the efficacy of FOS + meropenem (MER) remained indeterminate.[38]
E. coli2020, EgyptEl-Wafaimipenem8-3 (37.5%)7 (87.5%)in vitro (CB, TK)2 (25%)5 (62%)0%0%2 (66%)6 (87%)For 1 isolate the efficacy of FOS + MER remained indeterminate[42]
2019, IndiaSugathanmeropenem50-0%8 (16%)in vitro (TK)34 (68%)14 (28%)2 (4%)0%0% (all S)2 (25%)-[137]
2019, GermanyLoosemeropenem, ertapenem4-1 (25%)3 (75%)in vitro (CB)4 (100%)0%0%0%---[138]
2013, AustriaLingscheiddoripenem10ESBL (80%), AmpC (20%)0%-in vitro (CB, TK)8 (80%)-.0%--The authors reported FICI ranging from 0.5 to 4, without distinction between additive and indifferent effect.[139]
2012, GreeceSamonisimipenem, meropenem, doripenem20ESBL (100%)0%0%in vitro (ET)IMI: 11 (55%); MER: 5 (25%); DORI: 6 (30%)IMI: 9 (45%); MER: 15 (75%); DOR: 14 (70%)0%0%---[86]
2010, ThailandNetikulertapenem, imipenem, meropenem, doripenem8ESBL (87%)0%8 (100%)in vitro (ET)0%ERT: 5 (62%); IMI: 2 (25%); MER: 2 (25%); DOR: 1 (12%)ERT: 3 (37%); IMI: 6 (75%); MER: 6 (75%); DOR: 7 (87%)0%---[140]
K. pneumoniae2020, IndiaBakthavatchalammeropenem50OXA (78%), NDM (32%)-50 (100%)in vitro (TK)10 (20%)0%40 (80%)0%---[141]
2020, TurkeyErturk Sengelmeropenem17OXA (70%), NDM (70%) 7 (41%)17 (100%)in vitro (CB, TK)15 (88%)2 (11%)0%0%4 (23%)--[142]
2019, GermanyLoosemeropenem, ertapenem3-3 (100%)2 (66%)in vitro (CB)2 (66%)1 (33%)0%0%---[138]
2019, BrazilPerdigão Netomeropenem9ESBL, KPC (100%); OXA (4%), fosA (100%)9 (100%)9 (100%)in vitro (CB, TK)8 (88%)0%1 (11%)0%2 (22%)0%-[143]
2017, TaiwanTsengmeropenem25see comments12 (48%)24 (96%)in vitro (CB)25 (100%)0%0%0%--The 25 isolates were randomly selected among 642 isolates with the following resistance determinants: fosA3 (5.5%), foskp96 (4.2%), KPC (10.1%), IMP (0.8%), VIM (0.2%). It is not reported which carbapenemases and fosfomycinases were present in the 25 isolates tested for synergism.[144]
2017, ChinaYuimipenem, ertapenem136KPC (100%)78 (57%)136 (100%)in vitro (CB, TK)IMI: 21 (15%); ERT: 30 (22%)IMI: 114 (83%); ERT: 104 (76%)IMI: 1 (1%); ERT: 2 (1%)0%---[89]
2016, BrazilAlbieromeropenem18KPC (100%)13 (72%)16 (89%)in vitro (CB)12 (66%)3 (16%)3 (16%)0%12 (92.3%)4 (25%)-[145]
2014, SwedenTängdénmeropenem4NDM (50%), VIM (50%), ESBL (100%)2 (50%)3 (75%)in vitro (TK)0%0%4 (100%)0%---[146]
2013, TurkeyEvrenimipenem, meropenem12OXA-48 (100%)12 (100%)12 (100%)in vitro (CB)IMI: 5 (41%); MER: 4 (33%)IMI: 6 (50%); MER: 6 (50%)IMI: 1 (8%); MER: 2 (16%)0%---[74]
2013, AustriaLingscheiddoripenem5ESBL (60%), AmpC (100%)0%-in vitro (CB, TK)5 (100%)0%0%0%---[139]
2012, GreeceSamonisimipenem, meropenem, doripenem64KPC (78%), ESBL (21%)1 (1%)51 (78% )in vitro (ET)KPC: IMI: 37 (74%); MER: 35 (70%); DOR: 37 (74%). ESBL: IMI: 11 (78%); MER: 6 (42%); DOR: 6 (42%)KPC: IMI: 13 (26%); MER: 15 (30%); DOR: 13 (26%). ESBL: IMI: 3 (21%); MER: 8 (57%); DOR: 8 (57%)0%0%---[86]
2011, GreeceSoulimeropenem17KPC (100%)4 (23%)17 (100%)in vitro (TK)11 (64%)0%6 (35%)0%---[53]
2010, ThailandNetikulertapenem, imipenem, meropenem, doripenem8ESBL (87%)4 (50%)8 (100%)in vitro (ET)0%ERT: 5 (62%); IMI: 2 (25%); MER: 1 (12%); DOR: 2 (25%)ERT: 3 (37%); IMI: 6 (75%); MER: 7 (87%); DOR: 6 (75%)0%---[140]
E. cloacae2019, GermanyLoosemeropenem, ertapenem2-2 (100%)1 (50%)in vitro (CB)0%2 (100%)0%0%---[133]
2013, AustriaLingscheiddoripenem31 (33%)0%-in vitro (CB, TK)1 (33%)--0%--The authors reported FICI ranging from 0.5 to 4, without distinction between additive and indifferent effect.[139]
P. aeruginosa2020, USAMullanemeropenem30-14 (47%)30 (100%)in vitro (CB, TK)5 (17%)9 (30%)16 (53%)0%0%0%-[129]
2019, USAAverymeropenem153-NA (at least 71)153 (100%)in vitro (ET)29 (19%)55 (35%)69 (45%)0%-21 (13%)-[33]
2019, BrazilAlbieromeropenem19MBL (52%)17 (89%)16 (84%)in vitro (CB)15 (88%)3 (15%)1 (5%)0%15 (88%)7 (43%)-[147]
2019, USAFlammmeropenem5---in vitro (CB, TK)1 (20%)3 (60%)1 (20%)0%---[38]
2019, BrazilPerdigão Netomeropenem1OXA, fosA (100%)1 (100%)1 (100%)in vitro (CB, TK)1 (100%)0%0%0%1 (100%)1 (100%)-[143]
2018, USADrusanomeropenem1---in vitro (hollow-fiber infection model)1 (100%)0%0%0%--Combination therapy was able to counterselect resistance emergence.[148]
2017, SpainHamou-Segarraimipenem4-1 (25%)-in vitro (TK)4 (100%)0%0%0%--FOS and imipenem (IMI) alone lead to bacterial regrowth, while no regrowth was observed with the combination FOS + IMI.[149]
2015, ThailandKunakonvichayaimipenem, meropenem, doripenem70--70 (100%)in vitro (CB, TK)IMI: 38%; MER: 40%; DOR: 45%-----FOS in association with a carbapenem was observed to reduce also biofilm formation.[150]
2013, Brazildos Santosimipenem4-4 (100%)2 (50%)in vitro (CB)4 (100%)0%0%0%3 (75%)1 (50%)-[48]
2013, AustriaLingscheiddoripenem18---in vitro (CB, TK)0%0%18 (100%)0%--The authors reported FICI ranging from 0.5 to 4, without distinction between additive and indifferent effect, and considered the combination "indifferent" against all isolates.[139]
2012, GreeceSamonisimipenem, meropenem, doripenem15-1 (1%)9 (60%)in vitro (ET)IMI: 7 (46%); MER: 8 (53%); DOR: 11 (73%)IMI: 8 (53%); MER: 7 (46%); DOR: 4 (26%)0%0%---[86]
2005, ThailandPruekprasertimipenem29---in vitro (CB)11 (38%)4 (14%)12 (41%)2 (7%)---[22]
2002, JapanOkazakiimipenem, meropenem30-15 (50%)IMI: 29 (96%); MER: 27 (90%)in vitro (efficacy time index)IMI: 22 (73%); MER: 26 (86%)IMI: 0%; MER: 2 (6%)IMI: 8 (26%); MER: 2 (6%)0%IMI: 2 (13%); MER: 3 (20%)IMI: 21 (72%); MER: 16 (59%)-[39]
1999, JapanHayamimeropenem26-NA (at least 13)NA (at least 5)in vitro (CB, TK)3 (11%)15 (57%)8 (30%)0%---[130]
1997, FranceTessierimipenem40-20 (50%)9 (22%)in vitro (CB)0%15 (37%)25 (62%)0%17 (85%)8 (88%)Although the combination had a synergistic effect on no tested strains, it is of clinical relevance as it restored FOS and IMI susceptibility in almost all R isolates.[131]
A. baumannii2019, USAFlammmeropenem5 (A. baumannii-calcoaceticus species complex)---in vitro (CB, TK)1 (20%)3 (60%)0%0%--For 1 isolate the efficacy of FOS + MER remained indeterminate.[38]
2018, ChinaZhuimipenem21-20 (95%)21 (100%)in vitro (CB)12 (57%)3 (14.3%)6 (28%)0%---[151]
2018, ThailandSingkham-Inimipenem, meropenem23OXA (100%)23 (100%)23 (100%)in vitro (CB, TK)IMI: 65%; MER: 0%IMI: 30.4%; MER: 87%IMI: 4%; MER: 13%0%---[152]
2016, BrazilLeiteimipenem, meropenem20OXA (100%), IMP (15%)20 (100%)20 (100%)in vitro (CB, TK)IMI: 0%; MER: 0%IMI: 4 (20%); MER: 0%IMI: 16 (80%); MER: 100%0%---[83]
1996, SpainMartinez-Martinezimipenem34-34 (100%)NA (at least 7)in vitro (CB)1 (3%)--0%--The Authors reported only the number of isolates on which the combination had a synergistic or an antagonistic effect.[132]
S. aureus2019, SpainCoronado-Álvarezimipenem4MRSA (50%)--in vitro (TK)4 (100%)0%0%0%---[63]
2015, Spaindel Ríoimipenem10 (in vitro); 2 (in vivo)MRSA (100%)1 (10%)4 (40%)in vitro (TB); in vivo (rabbit, endocarditis)in vitro: 9 (90%); in vivo: 2 (100%)in vitro: 1 (10%)0%0%--% of sterile vegetations: FOS alone 0%, IMI alone 7%, FOS + IMI 73%.[28]
2013, AustriaLingscheiddoripenem39MRSA (100%)0%-in vitro (CB, TK)37 (94%)--0%--The authors reported FICI ranging from 0.5 to 4, without distinction between additive and indifferent effect.[139]
2012, SpainGarrigósimipenem1MRSA (100%)0%0%in vitro (TK); in vivo (rat, foreign-body infection)0%in vitro: 1 (100%)in vitro: 0%; in vivo: 1 (100%)0%---[37]
2011, SpainPachón-Ibáñezimipenem1GISA (100%)0%100%in vitro (TK); in vivo (mouse, peritonitis)in vitro: 1 (100%); in vivo: 1 (100%)0%0%0%--FOS + IMI reached statistical difference when compared to IMI as single therapy in the mouse model.[36]
2003, JapanNakazawaimipenem, panipenem32MRSA (100%)29 (91%)28 (88%)in vitro (efficacy time index)IMI: 16 (50%); PAN: 21 (66%)IMI: 3 (9%); PAN: 8 (25%)IMI: 13 (41%); PAN: 3 (9%)0%---[18]
1987, FranceQuentinimipenem5-1 (20%)1 (20%)in vitro (TK)1 (20%)0%3 (60%)1 (20%)---[35]
S. aureus + S. epidermidis2001, AustriaGrifmeropenem5 S. aureus + 2 S. epidermidisMRSA (25%), GISA (25%)--in vitro (CB, TK)S. aureus: 5 (100%)0%S. epidermidis: 2 (100%)0%---[43]
1992, AustriaGuggenbichlerimipenem1 S. aureus + 2 S. epidermidis---in vitro (TK)3 (100%)0%0%0%--The study was conducted on catheters infected in laboratory. Bacterial regrowth was observed in catheters treated with FOS or IMI alone, but did not occurred when the drugs were tested in combination.[153]
Staphylococcus spp. + Enterococcus spp.1986, ItalyDebbiaimipenem76---in vitro (CB, TK)54 (71%)0%22 (29%)0%--% reported are those obtained with CB. Results of TK showed higher rates of synergism, but in the present Table are considered the results of CB as not all isolates were tested with TK. [154]
E. faecalis2011, ItalyFarinaimipenem27-2 (7%)0%in vitro (ET)0%0%10 (37%)17 (62%)--The Authors did not distinguish between additive and indifferent effect, and defined the effect of FOS + IMI indifferent.[128]
S. pneumoniae1994, FranceDoitimipenem26-0%0%in vitro (TK)0%26 (100%)0%0%---[134]
N. gonorrhoeae2015, The NetherlandsWindertapenem4---in vitro (ET)0%3 (75%)1 (25%)0%---[54]
Table 4. Studies on combination between fosfomycin and aztreonam. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 4. Studies on combination between fosfomycin and aztreonam. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Aztreonam-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%) Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Aztreonam Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAAvery4848 not specified between: 8 E. coli: KPC (25%), NDM (75%), ESBL (62%); 35 Klebsiella spp: KPC (45%), NDM (40%); OXA (14%), VIM (8.%), ESBL (88%), fosA (44%); 2 Citrobacter spp: KPC (50%), NDM (50%), ESBL (50%), 4 E. cloacae: KPC (75%), NDM (25%), ESBL (75%)20 (40%)48 (100%)in vitro (ET)4 (8%)13 (27%)31 (64%)0%0%0%Data on synergism reported without distinction for bacterial strains. % of FOS-R isolates estimated on the basis of the reported MIC50.[11]
2019, USAFlamm20---in vitro (CB, TK)5 (25%)5 (25%)1 (5%)0%--For 9 isolates the efficacy of FOS + ATM remained indeterminate.[38]
E. coli2014, SwedenHickam2ESBL, OXA (50%)0%1 (50%)in vitro (CB, TK)2 (100%)0%0%0%---[120]
K. pneumoniae2014, SwedenHickam1ESBL, OXA (100%)0%1 (100%)in vitro (CB, TK)0%1 (100%)0%0%---[120]
P. aeruginosa2019, USAAvery103-NA (at least 71)103 (100%)in vitro (ET)16 (15.5%)68 (66%)19 (18%)0%-21 (13%)-[33]
2019, USAFlamm5---in vitro (ET)1 (20%)3 (60%)0%0%--For 1 isolate the efficacy of FOS + ATM remained indeterminate.[38]
2002, JapanOkazaki30-15 (50%)29 (96%)in vitro (efficacy time index)23 (76.%)3 (10%)4 (13%)0%4 (26%)6 (20%)-[39]
Table 5. Studies on combination between fosfomycin and quinolones. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 5. Studies on combination between fosfomycin and quinolones. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorQuinoloneNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Quinolone-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Quinolone Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAFlammLevofloxacin207 MDR (of which 29% ESBL and 29% KPC-producer)--in vitro (CB)30%60%10%0%---[38]
E. coli2020, EgyptEl-WafaCiprofloxacin8-100%100%in vitro (CB, TK)3 (37%)---3 (100%)3 (100%)Triple combination (FOS/IMP/CIP o FOS/CIP/TOB) increased synergism against all isolates.[42]
2019, USAWangCiprofloxacin8-25%25%in vitro (ET, biofilm)2 (25%)-6 (75%)-0%0%-[155]
2019, IndiaSugathanCiprofloxacin50biofilm producers (100%)0%98%in vitro (CB, TK)3 (6%)20 (40%)27 (54%)0%-0%The optimal combination of fosfomycin with N-acetylcystein produces the reduction of E. coli sessile cell viability and biofilm formation up to 60–73%.[137]
S. flexneri2019, ChinaLiuCiprofloxacin80-43 (54%)100%in vitro (CB, TK); in vivo (Galleria mellonella)31 (38%)0%49 (61)%0%65 (81%)3 (4%)-[156]
P. aeruginosa2019, USAWangCiprofloxacin7-0%14%in vitro (ET, biofilm)4 (57%)-3 (42%)--0%-[155]
2019, USAFlammLevofloxacin57 MDR (of which 29% ESBL and 29% KPC-producer)--in vitro (CB)1 (20%)4 (80%) 0%---[38]
2016, AustraliaWalshCiprofloxacin4-75%50%in vitro (TK)21% (23/108)15% (16/108)38% (41/108)---The total number of experiments was 108 (9 combinations of FOS + CIP at different concentrations, in 3 different times).[76]
2013, BrazilDos SantosCiprofloxacin2MDR (50%)100%50%in vitro (CB, TK)2 (100%)---2 (100%)0%-[48]
2007, JapanMikuniyaPrulifloxacin, ciprofloxacin, levofloxacin1biofilm forming (100%)--in vivo (rat, UTI)1 (100%)-----*After 3 consecutive days’ co-administration.[40]
2007, JapanYamadaCiprofloxacin74---in vitro (CB)20 (27%)-54 (73%)0%---[157]
2005, JapanMicuniyaCiprofloxacin, Ulifloxacin, Levofloxacin1-100%100%in vitro (ATP bioluminescence assay)-100%--0%0%-[46]
2002, JapanMondenOfloxacin4-3 (75%)1 (25%)in vitro (biofilm)3 (75%)------[158]
2001, JapanOkazakiLevofloxacin30MDR (50%)13/30 (43%)21/30 (70%)in vitro (Efficacy time index)3/30 (1%)17/30 (56%)-10/30 (33)%--ETI < 0.5 antagonism; 0.5 ≤ ETI <1 indifferent; 1 ≤ ETI < 8 additive; ETI ≥ 8 synergistic[39]
1999, JapanHayamiCiprofloxacin26---in vitro (CB, TK)10(38%)15 (57%)1 (3%)0%---[130]
1997, FranceBugnonPefloxacin2---in vivo (rabbit, endocarditis)---100%---[41]
1997, FranceTessierCiprofloxacin40MDR (100%)23 (57%)19 (47%)in vitro (CB)6 (15%)32 (80%)2 (5%)-16 (70%)12 (63%)-[131]
1995, JapanKumonOfloxacin1---in vitro (TK)1 (100%)------[159]
1994, FranceXiongCiprofloxacin2MDR (50%)0%50%in vitro (CB); in vivo (rabbit, endocarditis)2 (100%) early thp; 1 (50%) Late thp0% early thp; 1 (50%) Late thp----in vivo results.[160]
1994, FranceXiongPefloxacin2MDR (50%)0%50%in vitro (CB); in vivo (rabbit, endocarditis)1 (50%) early thp; 1 (50%) late thp1 (50%) early thp1 (50%) late thp---in vivo results.[160]
1989, GermanyVogtCiprofloxacin25-1 (4%)2 (8%)in vitro (TK)20%------[161]
1988, USAFigueredoCiprofloxacin----in vitro (CB)60% (EV) 17% (OS)--0%---[162]
1987, GermanyUllmannCiprofloxacin37---in vitro (CB)29 (78%)8 (22%)0%0%100%--[45]
A. baumannii1996, SpainMartinez-MartinezCiprofloxacin34-100%100%in vitro (CB)1 (3%)--0%---[132]
A. baumannii-A. calcoaceticus spp. complex2019, USAFlammLevofloxacin57 MDR (29% ESBL and 29% KPC-producer)--in vitro (CB)0%4 (80%)1 (20%)0%---[38]
Gram negative1977, SpainDazaNalidixic acid100-100%-in vitro (CB)0%-100%0%--Klebsiella spp., Pseudomonas spp., E. coli, Serratia spp., Proteus spp., Enterobacter spp., Acinetobacter spp., Levinea spp.[66]
Staphylococcus spp.2003, JapanNakazawaOfloxacin32MRSA (100%)--in vitro (efficacy time index)3 (9%)2 (6%)27 (84%)---synergism = high efficacy; additive = efficacy; indifferent = invalid[18]
2001, AustriaGrifMoxifloxacin7MRSA (100%)--in vitro (CB)100%------[43]
1997, ItalyFerraraSparfloxacin16MRSA (100%)>50%∼100%in vitro (TK)0%------[123]
1988, FranceThauvinPefloxacin1MRSA (100%) in vivo (rat, endocarditis)100%------[44]
1987, FranceWeberOfloxacin8MRSA (37%)--in vitro (TK)2 (25%)6 (75%)-----[163]
1987, GermanyUllmannCiprofloxacin20---in vitro (CB)19 (95%)1 (5%)----S. aureus. [45]
1987, FranceQuentinPefloxacin6-16%0%in vitro (TK)0%0%100%0%--S. aureus. Indifferent effect.[35]
S.epidermidis1997, ItalyFerraraSparfloxacin12MRSE (100%)>50%∼100%in vitro (TK)6/12 (50%)------[123]
1987, FranceQuentinPefloxacin2-50%-in vitro (TK)0%0%100%0%--Indifferent effect.[35]
N. gonorrhoeae2014, NetherlandsWindMoxifloxacin4---in vitro (ET)0%------[54]
Table 6. Studies on combination between fosfomycin and aminoglycosides. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 6. Studies on combination between fosfomycin and aminoglycosides. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorAminoglycosideNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Aminoglycoside-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Aminoglycoside Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAAveryTobramycin4545 not specified between: 8 E. coli: KPC (25%), NDM (75%), ESBL (62%); 35 Klebsiella spp: KPC (45%), NDM (40%); OXA (14%), VIM (8%), ESBL (88%), fosA (44%); 2 Citrobacter spp: KPC (50%), NDM (50%), ESBL (50%), 4 E. cloacae: KPC (75%), NDM (25%), ESBL (75%)20/49 (40%)45 (100%)in vitro (ET)2 (4%)7 (15%)36 (80%)0%--Data on synergism reported without distinction for bacterial strains. Percentages of FOS-R isolates estimated on the basis of the reported MIC50.[11]
2019, USAFlammGentamicin20---in vitro (CB, TK)6 (30%)13 (65%)1 (5%)0%---[38]
1978, SpainOlayStreptomycin, gentamicin, kanamycinStreptomycin: 18 E. coli. Gentamicin: 30 E. coli, 24 Klebsiella spp., 39 S. marcescens, 33 Proteus spp. Kanamycin: 21 E. coli, 12 Klebsiella spp., 16 Proteus spp., 5 E. cloacae, 22 S. marcescens---in vitro (CB)streptomycin: 0%; gentamicin: 16 (12%); kanamycin: 21 (27%)streptomycin: 9 (50%); gentamicin: 52 (41%); kanamycin: 37 (48%)streptomycin: 9 (50%); gentamicin: 58 (46%); kanamycin: 18 (23%)0%---[14]
E. coli2020, EgyptEl-Wafatobramycin8-3 (37.5%)8 (100%)in vitro (CB, TK)2 (25%)0%0%0%2 (66%)2 (25%)For 6 isolates the efficacy of FOS + TOB remained indeterminate.[42]
2019, USAWangGentamicin8-0%2/8 (25%)in vitro (ET, biofilm)75% (6/8)0%(2/8) 25%0%-1/2 50%-[155]
2019, IndiaSugathanAmikacin50-0%26 (52%)in vitro (TK)29 (58%)21 (42%)0%0%0% (all S)22 (84%)The Authors also studied the efficacy of combination of FOS + AMK and found it reduced significantly biofilm formation.[137]
2013, SwitzerlandCorvecGentamicin1CTX-M15, ESBL0%0%in vitro (TK); in vivo (foreign-body infection model)0%100%0%0%-- Cure rate of FOS plus gentamicin 42%.[73]
2011, GreeceSamonisNetilmicin20ESBL0%35%in vitro (ET)25% (5/20)------[86]
1977, PolandBorowskiStreptomycin10---in vitro (CB)7 (70%)3 (30%)0%0%---[121]
K. pneumoniae2020, TurkeyErturk SengelAmikacin17OXA-48, NDM41%76%in vitro (CB)29%29%24%0%--Combination of FOS plus amikacin seems not a good choice for NDM producing strains.[142]
2018, ChinaYuAmikacin3-0%-in vitro (TK)100% (3/3)0%0%0%---[164]
2017, ChinaYuAmikacin3KPC-20%33%in vitro (TK)66%0%33%0%--FOS (8 g q8h)/AMK (15 mg/kg qd) most bactericidal activity, but resistance occurred.[50]
2017, ChinaYuAmikacin136KPC (100%)78 (57%)80 (58%)in vitro (CB, TK)7 (5%)109 (80%)20 (14%)0%---[89]
2015, SpainRodriguez-Avial et al.Plazomicin4 (CB); 2 (TK)Carbapenemase-producing strains (KPC, VIM)100%NAin vitro (CB, TK)25–100%50–0%25–0%0%---[51]
2014, USAMontgomeryAmikacyn20KPC-2 (20%), KPC-3 (15%)-100%in vitro (agar diluition, antibiotic potentation study in A:F 5:2 ratio)100%--0%--Synergy defined: reduction of FOS and AMK MIC when used in combination.[52]
2011, GreeceSamonisNetilmicin65serine carbapenem-producing (50/65); ESBL (14/65); MBL (1/65)98%87%in vitro (ET)41% (27/65) overall. In ESBL 42% (6/14). In serine enzymes 42% (21/50)----54% (25/46)-[86]
2011, GreeceSouligentamicin17KPC (100%)4 (23%)7 (41%)in vitro (TK)0%0%15/15 (100%)---Efficacy of FOS + GEN was not evaluated in 2 isolates.[53]
1977, SpainDazaTobramicin23---in vitro (CB)2/23 (8%)--0%---[66]
M. morganii1977, SpainDazaGentamicin2---in vitro (CB)50% (1/2)--0%---[66]
P. aeruginosa2019, USAWangGentamicin7-25%1/7 (14%)in vitro (ET, biofilm)4 (57%)0%3 (42%)0%-0%-[155]
2019, USAAverytobramycin42-NA (at least 71)42 (27%)in vitro (ET)8 (19%)13 (31%)21 (50%)0%-8 (19%)-[33]
2019, New ZealandLi BassiAmikacin15Strains resistant to nebulized fosfomycin and amikacin (100%)--in vivo (pigs, pneumonia)0%0%100%0%--No difference in P. aeruginosa lung tissue concentration, bronchoalveolar lavage concentration and lung hystopathology score when amikacin and FOS were administered by aerosol alone or in combination therapy.[165]
2019, USAFlammgentamicin, amikacin5---in vitro (CB, TK)0%genta: 4 (80%); amika: 4 (80%)genta: 1 (20%); amika: 1 (20%)0%---[38]
2018, SpainDiez-AguilarTobramycin6-100%67%in vitro (CB)83%17%0%0%--Synergy tested in biofilm.[166]
2015, AustraliaWalshTobramycin3- 1/4 (25%)in vitro (TK)18% (15/81)25% (20/81)-----[76]
2015, SpainDiez-AguilarTobramycin8mexZ mutation (25%), ANT(2’)-I enzyme (37.5%),100%37%in vitro (TK)25%0%75%0%---[166]
2014, USAMontgomeryAmikacin21GES-1, OXA-2 plus OXA-10 plus VIM-2, OXA 14, VIM-4 (each, 4.8%), VIM-2 (19%)-100%in vitro (agar dilution, antibiotic potentation study in A:F 5:2 ratio)100%-----Synergy defined: reduction of FOS and AMK MIC when used in combination.[52]
2013, BrazilFerrari dos Santos LimaTobramycin2IMP-R (100%)100%100%in vitro (broth microdilution, CB)100%0%0%0%100%0%Authors do NOT report FOS and AMG MIC (they referred to CLSI criteria except for FOS-Eucast S ≤ 32 µg/mL); FOS MIC restoration 32.[48]
2013, USAAndersonTobramycin1---in vitro (effects on biofilms on CF airway epithelial cells)-100%0%0%--FOS:TOBRA (4:1) formulas for inhalation treatment; results suggest that fosfomicon enhanced the activity of tobramycin (much less level of tobramycin needed). FOS alone does NOT result in biofilm inhibition, TOBRA alone require HIGHER doses for biofilm inhibition.[49]
2012, UK/USAMcCaugheyTobramycin15---in vitro (agar dilution, TK)100%--0%--Synergism defined as FOS:TOBRA bactericidal activity; Time kill studies in a subset of isolates; biofilm studies were also performed.[167]
2011, GreeceSamonisNetilmicin15MDR93%13%in vitro (ET)13% (2/15)------[86]
2009, ChinaCaiAmikacin20--NA (MIC90 32)in vitro (CB); in vivo (rat, biofilm-infected model)80%15%-0%-MIC90 decrease of 64-foldF + T (lowest FICI amikacina and isepamicina) had synergistic effect on planctonic P. aeruginosa.[168]
2009, ChinaCaiGentamicin20--NA (MIC90 16)in vitro (CB); in vivo (rat, biofilm-infected model)70%15%-0%-MIC90 decrease of 8-foldF + T (lowest FICI amikacina and isepamicina) had synergistic effect on planctonic P. aeruginosa.[168]
2009, ChinaCaiNetilmicin20--NA (MIC90 16)in vitro (CB); in vivo (rat, biofilm-infected model)65%20%-0%-MIC90 decrease of 8-foldF + T (lowest FICI amikacina and isepamicina) had synergistic effect on planctonic P. aeruginosa.[168]
2009, ChinaCaiTobramycin20--NA (MIC90 8)in vitro (CB); in vivo (rat, biofilm-infected model)60%20%-0%-MIC90 decrease of 2-foldF + T (lowest FICI amikacina and isepamicina) had synergistic effect on planctonic P. aeruginosa.[168]
2005, ThailandPruekprasertgentamicin22---in vitro (CB)1 (4%)9 (42%)6 (27%)6 (27%)---[22]
2002, JapanOkazakigentamicin30-15 (50%)19 (63%)in vitro (efficacy time index)0%9 (30%)21 (70%)0%0%15 (50%)-[39]
1999, JapanHayamiamikacin26-NA (at least 13)NA (< 5)in vitro (CB, TK)0%10 (38%)16 (61%)0%---[130]
1991, NigeriaChinwubaGentamicin8--0%in vitro (CB, TK)0%0%100%0%--- [169]
1997, FranceTessieramikacin40-23 (57%)13 (32%)in vitro (CB)3 (7%)21 (52%)16 (40%)0%18 (78%)11 (84%)Although the combination had a synergistic effect on no tested strains, it is of clinical relevance as it restored FOS and AMK susceptibility in many resistant strains.[131]
1978, SpainOlaygentamicin, kanamycin77 gentamicin, 15 kanamycin---in vitro (CB)gentamicin: 55 (71%); kanamycin: 4 (26%)gentamicin: 17 (22%); kanamycin: 8 (53%)gentamicin: 5 (6%); kanamycin: 3 (20%)0%---[14]
A. baumannii2019, USAFlammgentamicin, amikacin5 (A. baumannii-calcoaceticus species complex)---in vitro (CB, TK)genta: 2 (40%); amika: 2 (40%)genta: 3 (60%); amika: 3 (60%)0%0%---[38]
2016, BrazilLeitegentamicin, amikacin20OXA (100%), IMP (15%)20 (100%)genta: 11 (55%); amika: 19 (95%)in vitro (CB, TK)0%genta: 2 (10%); amika: 0%genta: 18 (90%); amika: 20 (100%)0%--"2-well" method showed synergistic activity in about 20% of tested strain, but the Authors considered it not fully reliable and concluded the association had an indifferent effect.[83]
2014, USAMontgomeryAmikacyn21OXA-23 plus OXA-51 (23.8%); OXA-24 plus OXA-51 (9.5%), OXA-51, OXA-51 plus OXA-58 (each, 4.8%)-100%in vitro (agar dilution, antibiotic potentation study in A:F 5:2 ratio)100%--0%--Synergism defined as reduction of FOS and AMK MIC when used in combination.[52]
1996, SpainMartinez-Martinezamikacin, tobramycin34-34 (100%)amika: 31 (91%); tobra: 33 (97.%)in vitro (CB)amika: 15 (44%); tobra: 11 (32%)--0%--The authors reported only synergistic and antagonistic effect rates.[132]
Gram-negative1977, SpainDazaTobramycin75---in vitro (CB)0%0%100%0%--33 Klebsiella spp., 21 P. aeruginosa, 3 P. cepacia, 12 E.coli,11 S. marcescens, 9 Enterobacter spp., 8 Proteus spp., 2 A. calcoaceticus, 1 L. malonatica, 5 K. pneumoniae oxytoca, 5 K. Ozenae, 5 E. aerogenes, 2 E. hafniae, 1 E. cloacae, 1 E. liquefaciens, 4 P. mirabilis, 2 P. rettgeri[66]
1977, SpainDazaGentamicin75---in vitro (CB)0%0%100%0%--[66]
S. aureus2017, SpainLopez DiazPlazomicin12 (BC); 5 (TK)MRSA Strains carrying aminoglycosides-modifying enzymes (100%)56%-in vitro (CB, TK)33.3–0%66–100%0%0%---[170]
2012, UK/USAMcCaugheyTobramycin5MRSA100%-in vitro (agar dilution, TK)60%--0%--Synergism defined as F:T bactericidal activity; Time kill studies in a subset of isolates; biofilm studies were also performed[167]
2005, JapanMorikawaArbekacin1MRSA100%100% MIC 0.5 (no available breakpoint)in vivo (rat, carboxymethyl cellulose pouch infection model)100%-----NOT available arbikacin EUCAST breakpoints; Synergistic effect was evaluated by i) morphological and histological studies showing dramatic change in biofilm and inflammatory response and by ii) decrease in the number of viable bacteria in vivo.[171]
1994, JapanKonoArbekacin96MRSA38%-in vitro66% (60/90)--0%--Better results of FOS-arbekacin combination in FOS susceptible strains.[172]
1987, SpainRodriguezGentamicin1MRSA0%0%in vivo (endocarditis in 10 rabbits)100% (1/1) 0% n. of rabbits’ death (0/10)0%0%0%---[61]
1985, USAAlvarezGentamicin148MRSA--in vitro (microtiter technique in a 1:1 ratio)(10/148) 7%0%90% (134/148)(4/148) 3%--Synergy was indicated if the MICs of both drugs decreased by at least one-fourth. If the MIC of one drug owed a fourfold or greater increase, it was assumed to be an indication of antagonism.[12]
1978, SpainOlaystreptomycin, gentamicin, kanamycin18 streptomycin, 29 gentamicin, 21 kanamycin---in vitro (CB)streptomycin: 1 (5%); gentamicin: 0%; kanamycin: 9 (43%)streptomycin: 10 (55%); gentamicin: 3 (10%); kanamycin: 7 (33%)streptomycin: 7 (38%); gentamicin: 26 (89%); kanamycin: 5 (23%)0%---[14]
Streptococcus spp.1978, SpainOlaystreptomycin16---in vitro (CB)0%9 (56%)7 (43%)0%---[14]
E. faecium2019, ThailandHemapanpairoaGentamicin8VRE (100%)100%13%in vitro (ET for FOS, broth microdilution for gentamicin)63%13%25%0%63%-Synergistic activity assessed as a fourfold reduction of MIC when FOS combined with gentamicin 1 mcg/mL.[55]
N. gonorrhoeae2015, The NetherlandsWindgentamicin4---in vitro (ET)0%1 (25%)3 (75%)0%---[54]
Miscellaneous 2009, USAMacLeodTobramycin27 (4 S. aureus, 17 P. aeruginosa, 5 E.coli, 1 H. influenzae)---in vitro (CB, TK); in vivo (rat, pneumonia)7% (1 P. aeruginosa, 1 E.coli)-93%0%--In vitro (agar plate dilution, broth microdilution, CB ON a SUBSET of ISOLATES, TK) and in vivo (rat bacterial pneumonia). NB: CB for 27 total strains: 4 S. aureus, 17 P. aeruginosa, 5 E. coli, 1 H. influenzae. FOS:TOBRA 4:1 was rapidly bactericidal and exhibited concentration -bactericidal killing in TK, with excellent activity against S. aureus and H. influenzae, but poor activity against S. maltophilia, B. cepacia; it was active against M. catarrhalis, E. coli, Klebsiella and S. pneumoniae.[173]
Table 7. Studies on combination between fosfomycin and macrolides. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 7. Studies on combination between fosfomycin and macrolides. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorMacrolideNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Macrolide-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%) Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Macrolide Susceptibility Restoration (%)CommentsReference
E. coli1978, SpainOlayErythromycin14---in vitro (CB)42%29%28%0%--Authors considered Synergistic effect when MIC of both antimicrobials was at least fourfold lower over initial MIC; partial synergy when MIC of one antimicrobials was at least fourfold lower and MIC of the other one 2 times lower over initial MIC; Indifferent effect when MIC of both antimicrobials was 2 times lower; antagonism when MIC of both increased 4 times over initial MIC. [14]
Klebsiella spp. 1978, SpainOlayErythromycin44---in vitro (CB)50%23%27%0%--Authors considered Synergistic effect when MIC of both antimicrobials was at least fourfold lower over initial MIC; partial synergy when MIC of one antimicrobials was at least fourfold lower and MIC of the other one 2 times lower over initial MIC; Indifferent effect when MIC of both antimicrobials was 2 times lower; antagonism when MIC of both increased 4 times over initial MIC. [14]
E. cloacae1978, SpainOlayErythromycin16---in vitro (CB)62%38%0%0%--Authors considered Synergistic effect when MIC of both antimicrobials was at least fourfold lower over initial MIC; partial synergy when MIC of one antimicrobials was at least fourfold lower and MIC of the other one 2 times lower over initial MIC; Indifferent effect when MIC of both antimicrobials was 2 times lower; antagonism when MIC of both increased 4 times over initial MIC. [14]
Proteus spp. (Indole +)1978, SpainOlayErythromycin13---in vitro (CB)53%46%0%0%--Authors considered Synergistic effect when MIC of both antimicrobials was at least fourfold lower over initial MIC; partial synergy when MIC of one antimicrobials was at least fourfold lower and MIC of the other one 2 times lower over initial MIC; Indifferent effect when MIC of both antimicrobials was 2 times lower; antagonism when MIC of both increased 4 times over initial MIC. [14]
P. aeruginosa1982, JapanKasaiMidecamycin2-0%2 (100%)in vitro (TK)/in vivo (Mice, peritonitis or subcutaneous infection)0%2 (100%)0%0%--In all in vivo experiment survival rates of mice that received MDM + FOS was statistically significant higher then when FOS or MDM were administrated alone, proving synergistic effect.[59]
1978, SpainOlayErythromycin29---in vitro (CB)38%59%3%0%--Authors considered Synergistic effect when MIC of both antimicrobials was at least fourfold lower over initial MIC; partial synergy when MIC of one antimicrobials was at least fourfold lower and MIC of the other one 2 times lower over initial MIC; Indifferent effect when MIC of both antimicrobials was 2 times lower; antagonism when MIC of both increased 4 times over initial MIC. [14]
S. aureus1978, SpainOlayErythromycin34---in vitro (CB)26%68%6%0%--Authors considered Synergistic effect when MIC of both antimicrobials was at least fourfold lower over initial MIC; partial synergy when MIC of one antimicrobials was at least fourfold lower and MIC of the other one 2 times lower over initial MIC; Indifferent effect when MIC of both antimicrobials was 2 times lower; antagonism when MIC of both increased 4 times over initial MIC. [14]
S. epidermidis2009, AustriaPresterlAzithromycin11-2 (18%)5 (45%)in vitro (Microtitre plate assay on Biofilm culture)------Combination of azithromycin with any of the tested antimicrobial agents did not reduce the biofilm ODr compared to the ODr of biofilms treated with single agents[58]
S. pseudointermedius2014, CanadaDiCiccoClarithromycin8MRSP (100%)5 (62%)8 (100%)in vitro (Microtitre plate assay)5 (62%)2 (25%)0%0%--FICI for 1 strains was reported as "Not available".[60]
Streptococcus spp. 1978, SpainOlayErythromycin26---in vitro (CB)15%27%57%0%--Authors considered Synergistic effect when MIC of both antimicrobials was at least fourfold lower over initial MIC; partial synergy when MIC of one antimicrobials was at least fourfold lower and MIC of the other one 2 times lower over initial MIC; Indifferent effect when MIC of both antimicrobials was 2 times lower; antagonism when MIC of both increased 4 times over initial MIC. [14]
N. gonorrhoeae2015, SwitzerlandHauserAzithromycin8 (4 TK)AZT-HLR (12,%)0%1 (12%)in vitro (CB, TK)CK: 0%; TK: 0%CK: 0%; TK: 0%CK: 8 (100%); TK: 4 (100%)CK: 0%; TK: 0%--Only 4 strains were tested with TKA. Authors used Enterobacterales FOS breakpoint as presumptive breakpoint for N. gonorrhoeae (EUCAST: S ≤ 32 mg/L; CLSI: S ≤ 64 mg/L).[57]
2015, NetherlandsWindAzithromycin4Azithromycin and Ceftriaxone Resistant (100%)--in vitro (ET)0%0%4 (100%)----[54]
Table 8. Studies on combination between fosfomycin and glycopeptides. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 8. Studies on combination between fosfomycin and glycopeptides. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorGlycopeptideNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Glycpeptide-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Glycopeptide Susceptibility Restoration (%)CommentsReference
A baumannii2016, BrazilLeiteVancomycin20OXA-23 (50%), OXA-143 (35%), IMP-type (15%), depletion of OMP 43 kDa (20%)19 (95%)Natural resistancein vitro (CB, TK)0%0%CB: 20 (100%)0%0%Breakpoints not availableTK showed indifference in all strains.[83]
S. aureus2018, ChinaXuVancomycin3-1 (33%)0%in vitro (CB)0%2 (66%)1 (33%)0%1 (100%)No resistant isolatesIn vitro concentrations - VAN (0.5, 1, 2 mg/L); FOS (32, 64 mg/L).[174]
2017, SpainCoronado-AlvarezVancomycin4Methicillin resistance (50%)--in vitro (TK)0%4 (100%)0%0%--The study also evaluated 15 patients with bacteremia caused by MRSA were treated with FOS in combination with VAN. Of these, 7 patients (46.7%) had negative blood cultures after 48 h of combination therapy.[63]
2012, TaiwanTangVancomycin, teicoplanin8Methicillin resistance (100%)2 (6%)VAN: 0%; TEC: 0%in vitro (TK)VAN: 8 (100%)0%TEC: 8 (100%)0%0%No resistant isolatesSynergistic concentrations were 64 mg/L for FOS and 2 mg/L for VAN, at 24 h. Indifference was detected with 8 mg/L for TEC at 24 h. Significant reduction of colony count in biofilm model when FOS was in combination with either VAN and TEC after 5 days.[69]
2011, TaiwanTangVancomycin5Methicillin resistance (100%)0%0%in vitro (TK)5 (100%)0%0%0%No resistant isolatesNo resistant isolatesAll strains had borderline MIC values for VAN (2 mg/L). In vitro synergistic concentrations were 2 mg/L for VAN and 64 mg/L for FOS.[175]
2010, SpainPachon-IbanezVancomycin1hGISA (100%)0%0%in vitro (TK); in vivo (mouse, peritonitis)1 (100%)0%0%0%No resistant isolateNo resistant isolateResistant (4 mg/L) sub-population frequency: 3.6 × 10−6 CFU/mL; in vitro synergistic concentrations were 1–2–4 mg/L for FOS and 1–2 mg/L for VAN at 24 h. In vivo combination was significant and effective in reducing bacteremia rates in 57% (n = 8 out of 14) of mice treated.[36]
2005, ItalyPistellaVancomycin, teicoplanin7Methicillin resistance (100%)5 (71%)VAN: 3 (42%); TEC: 6 (85.7%)in vitro (TK)VAN: 7 (100%); TEC: 0%VAN: 0%; TEC: 7 (100%)0%0%7 (100%)0%Synergistic concentrations were 8 mg/L for FOS and 1 × MIC for VAN (1, 2 or 4 mg/L respectively) at 24 h.[176]
1987, SpainRodriguezVancomycin1Methicillin resistance (100%)0%0%in vitro (TK); in vivo (rabbit, endocarditis)1 (100%)0%0%0%No resistant isolatesNo resistant isolatesIn vitro synergism at 24 and 48 h. Fixed concentrations of FOS at 8 mg/L and VAN at 1 mg/L. In vivo combination was successful in 10 rabbits (100%) showing sterile vegetations.[61]
1985, SpainAlvarezVancomycin148Methicillin resistance (100%)15 (10%)1 (1%)in vitro (CB)0%0%145 (98%)3 (2%)--1 strain was resistant to VAN (MIC > 32 mg/L).[12]
S. aureus, S. epidermidis2014, ChinaShiVancomycin3 (2 S. aureus, 1 S. epidermidis)Methicillin resistance (67%)3 (100%)0%in vitro (TK); in vivo (biofilm in rats’ tissues)3 (100%)0%0%0%0%No resistant isolatesIn vitro synergistic concentrations at 1 mg/L for VAN and 64 mg/L for FOS at 6h and 24 h. In vivo significative reduction of biofilm formation in rats’ tissues (4, 100%).[62]
2001, AustriaGrifVancomycin7 (5 S. aureus; 2 S. epidermidis)S. aureus: GISA 1 (20%), MRSA 1 (20%)-0%in vitro (CB, TK)0%0%CB: S. epidermidis 2 (100%); S. aureus 5 (71%)CB: S. epidermidis 0%; S. aureus 2 (28%)--TK showed indifference for all strains, with fixed concentration of FOS at 40 mg/L and VAN at 10 mg/L.[43]
1989, GermanyGatermannVancomycin33 (15 S. aureus; 18 S. epidermidis)---in vitro (CB)S. aureus: 1 (6%); S. epidermidis: 1 (5%)S. aureus: 8 (53%); S. epidermidis: 7 (39%)S. aureus: 6 (40%); S. epidermidis: 9 (50%)S. aureus: 0%; S. epidermidis: 1 (5%)--Synergistic concentrations not specified.[177]
E. faecalis, E. faecium, S. aureus, S. epidermidis, CONS1986, ItalyDebbiaTeicoplanin76 strains: 30 E. faecalis, 6 E. faecium, 20 S. aureus, 10 S. epidermidis, 10 CoNSMethicillin resistance (50% of S. aureus)--in vitro (CB, TK)CB: 20 (67%) E. faecalis; 4 (67%) E. faecium; 6 (60%) S. aureus; 6 (60%) MRSA; 1 (10%) S. epidermidis; 6 (60%) CONSCB: 10 (33%) E. faecalis; 2 (33%) E. faecium; 4 (40%) S. aureus; 4 (40%) MRSA; 9 (90%) S. epidermidis; 4 (40%) CONS0%0%--Synergistic concentrations not specified. 46 strains were tested also by TK. TK results-Synergism: 11 (92%) E. faecalis; 4 (100%) E. faecium; 6 (100%) S. aureus; 8 (100%) MRSA; 6 (75%) S. epidermidis; 8 (100%) CoNS. Additive effect: 1 (8%) E. faecalis; 2 (25%) S. epidermidis.[178]
S. pneumoniae2006, SpainRibesVancomycin2Resistance to penicillin (50%) and ceftriaxone (100%)0%0%in vitro (TK); in vivo (rabbit, menigitis)1 (50%)1 (50%)0%0%No resistant isolatesNo resistant isolatesIn vitro synergism at 24 h, at concentrations achievable in CSF. In vivo combination significant and effective in eradicating meningitis with sterile blood cultures (8, 100%).[24]
1994, FranceDoitVancomycin26Isolates not susceptible to penicillin (100%)0%0%in vitro (TK)0%0%100%0%No resistant isolatesNo resistant isolatesFixed concentrations of FOS at 40 mg/L and VAN at 3 mg/L.[134]
S. epidermidis1990, FranceGaillanrdVancomycin1-0%0%in vitro (TK)1 (100%)0%0%0%No resistant isolatesNo resistant isolatesSynergism at 4 h. Fixed concentrations of FOS at 12.5 mg/L and VAN at 7.5 mg/L. Effective to reduce biofilm formation (1; 100%).[179]
1990, GermanySimonVancomycin, teicoplanin20Methicillin resistant (100%)10 (50%)VAN: 0%; TEC: 2 (10%)in vitro (CB)VAN: 4 (20%); TEC: 9 (45%)VAN: 5 (25%); TEC: 6 (30%)VAN: 11 (55%); TEC: 5 (25%)VAN: 0%; TEC: 0%-VAN: no resistant isolates; TEC: NSSynergistic concentrations at 0.5 X MIC for FOS, TEC and VAN. Good efficacy in artificial biofilm model when isolates were fully susceptible to FOS.[180]
E. faecalis - E. faecium2013, TaiwanTangVancomycin, teicoplanin19 strains: 9 E. faecalis; 10 E. faeciumVancomycin resistant (100%)5 (55%) E. faecalis; 7 (70%) E. faeciumVAN: 19 (100%) both; TEC: 1 (11%) E. faecalis; 6 (60%) E. faeciumin vitro (TK)VAN: 3 (33%) E. faecalis, 3 (30%) E. faecium; TEC: 8 (89%) E. faecalis, 3 (30%) E. faecium0%VAN: 6 (67%) E. faecalis, 7 (70%) E. faecium; TEC: 1 (11%) E. faecalis, 7 (70%) E. faecium0%0%VAN: 3 (33%) E. faecalis; 3 (30%) E. faecium; TEC: 0%Synergistic concentrations were 64 mg/L for FOS, 4 mg/L for VAN and 8 mg/L for TEC, at 24 h. FOS-TEC had synergistic effect against biofilm-producing E. faecalis (4; 44%) and one E. faecium (1; 10%) isolates. FOS-VAN had synergistic effect against only one biofilm-producing E. faecalis isolate (1; 11%).[13]
Table 9. Studies on combination between fosfomycin and tetracyclines. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 9. Studies on combination between fosfomycin and tetracyclines. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorTetracyclineNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Tetracycline-Resistant (%)In Vitro (Methods)/In Vivo (Animal and site of Infection)Synergistic Effect (%) Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Tetracycline Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAFlammMinocycline207/30 MDR strains (A. baumannii, Enterobacterales e P. aeruginosa) included 2 ESBL e 2 KPC Enterobacterales--in vitro (CB)4 (20%)13 (65%)1 (5%)0%--Authors considered Partial Sinergy when FICI was between 0.5–1 and Additive effect for FICI = 1. Results for 2/20 strains (10%) were indeterminate.[38]
1977, SpainDazaTetracycline100-100 (100%)-in vitro (CB)2 (2%)-98%0%--Authors considered Synergistic effect when MIC was at least fourfold lower over initial MIC.[66]
P. aeruginosa2019, USAFlammMinocycline57/30 MDR strains (A. baumannii, Enterobacterales e P. aeruginosa)--in vitro (CB)2 (40%)3 (60%)0%0%--Authors considered Partial Sinergy when FICI was between 0.5–1 and Additive effect for FICI = 1.[38]
A. baumannii2013, ChinaZhangMinocycline25Pan-Drug-Resistant (100%)100%See Commentsin vitro (CB)12%56%32%0%0%100%Mean MIC for Minocycline was 16, MIC range 4-16. Authors used CLSI breakpoint for MIN (S ≤ 4 mg/L).[65]
S. aureus2012, TaiwanTangMinocycline33 (8 TK)MRSA (100%)6%61%in vitro (TK, Biofilm MTT-staining method)------Only 8 strains were tested with TK. Biofilm cultures were 94% MIN resistant and 94% FOS resistant. Cases of synergism were observed with FOS+MIN combination. Percentages or other data were not reported by authors. Combination of FOS + MIN determined a statistically significant reduction on ODRs in biofilm cultures compared to single drugs.[69]
2011, ChinaSunMinocycline87MRSA (100%)35 (40%)13 (14%)in vitro (CB)76 (87%)-11 (12%)0%100%92%Authors considered Indifferent effect for FICI between 0,5 and 4. CLSI breakpoint was used for MIN (S ≤ 4 mg/L) and E. faecalis FOS breakpoint as presumptive breakpoint for MRSA (S ≤ 64 mg/L).[70]
2003, JapanNakazawaMinocycline32MRSA (100%)29 (91%)26 (81%)in vitro (Efficacy Time Index)10 (31%)1 (3%)21 (65%)----[18]
E. faecalis2013, TaiwanTangMinocycline9VRE (100%)56%89%in vitro (TK, Biofilm Model)TKA: 2 (22%); BM: 1 (11%)-----Additive, Indifferent and antagonistic effect were not evaluated.[13]
E. faecium2013, TaiwanTangMinocycline10VRE (100%)70%80%in vitro (TK, Biofilm Model)TKA: 4 (40%); BM: 1 (10%)-----Additive, Indifferent and antagonistic effect were not evaluated.[13]
2012, USADescourouezMinocycline32VRE (100%)9%See Commentsin vitro (TK)0%0%100%0%--The authors considered MIC ≤ 64 mg/L as FOS breakpoint. Most of strains were minocycline resistant (MIC range 4–32, mean MIC 16 mg/L).[67]
2019, USADavisDoxycycline24VRE (100%)96%8%in vitro (ET, TK)CK: 11 (46%); TK: 10 (41%)CK: 13 (54%); TK: 4 (16%)CK: 0%; TK: 10 (41%)CK: 0%; TK 0%--Authors used CLSI breakpoint for DOX (S ≤ 4 mg/L) and E. faecalis FOS breakpoint as presumptive breakpoint for E. faecium (S ≤ 64 mg/L).[68]
N. gonorrhoeae2015, NetherlandsWindMinocycline4Azithromycin and Ceftriaxone Resistant (100%)--in vitro (ET)0%0%4 (100%)----[54]
Table 10. Studies on combination between fosfomycin and polymyxins. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 10. Studies on combination between fosfomycin and polymyxins. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorPolymyxinNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Polymyxin-ResIstant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%) Additive Effect (%) Indifferent Effect (%) Antagonistic Effect (%) FOS Susceptibility Restoration (%)Polymyxin Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAFlammColistin20carbapenem-resistant (5%), KPC (10%), ESBL (10%)--in vitro (CB, TK) 1 (5%)5 (25%)8 (40%)0%--For 6 isolates the effect of the combination was indeterminate.[38]
2015, UKAlburColistin6NDM-1 (100%)3 (50%)0%in vitro (TK)3 (50%)0%3 (50%)0%--The combination was synergistic against FOS-S isolates. Against FOS-R isolates, an additive effect was observed after 12h, but then regrowth occurred.[181]
E. coli2013, SwitzerlandCorvecColistin1CTX-M15, ESBL (100%)0%0%in vitro (TK), in vivo (foreign-body infection model)1 (100%)0%0%0%---[73]
2011, FranceBerçotColistin1NDM-10%0%in vitro (CB, TK)0%1 (100%)0%0%--E. coli J53[85]
2011, GreeceSamonis Colistin20ESBL (100%)0%0%in vitro (ET)3 (15%)--0%---[86]
E. cloacae2011, FranceBerçotColistin2NMD-11 (50%)0%in vitro (CB, TK)0%2 (100%)0%0%---[85]
K. pneumoniae2020, TurkeyBuket Erturk Sengel Colistin17KPC (OXA-48, NDM) (100%)41%65%in vitro (CB, TK)7 (41%)3 (18%)5 (29%)2 (12%)---[142]
2019, IndiaBakthavatchalamColistin50CR-Kp, NDM, OXA-43 (100%)24 (48%)14 (30%)in vitro (TK)8 (16%)0%42 (84%)0%---[141]
2020, SwedenWistrand-YuenPolymyxin B5 (4 used for FOS+PMB)KPC-2, KPC-3, NMD-1, OXA-48, VIM-1 (100%)3 (60%)2 (40%)in vitro (TK)5 (31%)2 (12%)----Synergistic rate inferred from 4 isolates monitored at different times. If evaluated only after 24 h, syn: 40%; add 20%.[182]
2019, FranceCrémieuxColistin1carbapenem-resistant (100%)0%0%in vitro (TK); in vivo (rabbit, osteomyelitis)1 (100%)0%0%0%---[71]
2018, China Wang Colistin4carbapenem-resistant (100%)2 (50%)0% (75% heteroresistant)in vitro (TK)31 (43%)8 (11%)33 (46%)0%--3 isolates showed heteroresistance: the total number of experiments was 72 (3 different colistin concentrations tested in 6 different times).[183]
2018, China Yu Colistin3KPC (100%)1 (33%)3 (100%)in vitro (TK)2 (66%)1 (33%)0%0%---[164]
2017, TaiwanKuColistin9ESBL-producing KP (5/9 carbapenem-R, 4/9 carbapenem-S)4 (45%)1 (11%)in vitro (TK)5 (55%)0%4 (45%)0%---[84]
2017, ChinaYu Colistin3KPC2 (100%)0%1 (33%)in vitro (TK)3 (100%)0%0%0%---[50]
2017, ChinaYu Colistin136KPC-Kp (100%)78 (57%)1 (1%)in vitro (CB, TK)5 (3%)109 (80%)22 (16%)0%---[89]
2018, USABulmanPolymyxin B2KPC-2 (100%)0%0%in vitro (TK); in vivo (hollow-fibre infection model)2 (100%)------[75]
2014, SwedenTängdeén Colistin4VIM (50%), NDM (50%)2 (50%)0%in vitro (TK)3 (75%)0%1 (25%)0%--Synergism in 1 VIM- and 2 NDM-producing isolates, although NDM-producing isolates were FOS-R.[146]
2013, TurkeyEvrenColistin12OXA-48 (100%)11 (92%)2 (17%)in vitro (CB)0%0%0%12 (100%)---[74]
2011, FranceBerçotColistin3NDM-1 (100%)0%0%in vitro (CB, TK)0%1 (33%)2 (66%)0%---[85]
2011, GreeceSamonis Colistin50carbapenem-resistant (100%)3%25%in vitro (ET)18 (36%)--0%---[86]
2011, GreeceSamonis Colistin14ESBL (100%)3%25%in vitro (ET)1 (7%)--0%---[86]
2011, GreeceSouliColistin17KPC-2 (100%)4 (23%)7 (41%)in vitro (TK)2 (12%)0%15 (88%)0%---[53]
K. oxytoca2011, FranceBerçotColistin1NDM-10%0%in vitro (CB, TK)0%100%0%0%---[85]
P. rettgeri2011, FranceBerçotColistin1NDM-10%100%in vitro (CB, TK)0%0%100%0%---[85]
P. aeruginosa2019, USAFlamm Colistin5---in vitro (CB, TK) 0%1 (20%)4 (80%)0%---[38]
2016, AustraliaWalsh Polymyxin B4MDR (75%)50%50%in vitro (TK) 19 (18%)27 (25%)----FOS in combination with polymyxin B increased bacterial killing, but did not suppress emergence of FOS resistance. The total number of experiments was 108 (9 combinations of FOS + CIP at different concentrations, in 3 different times).[76]
2011, GreeceSamonis Colistin15MDR (100%)6%0%in vitro (ET) 2 (13%)--0%---[86]
2015, ChinaDiColistin87CRPA (100%)75%4% (5/87)in vitro (CB, TK) 19 (21%)29 (33)%39 (44%)0%-3 (60%)-[184]
A. baumannii-A. calcoaceticus spp. Complex2019, USAFlammColistin5MDR (20%)--in vitro (CB, TK) 2 (40%)1 (20%)1 (20%)0%--For 1 isolate the effect of the combination was indeterminate.[38]
A. baumannii2020, South KoreaSu KuColistin1OXA-23 (100%)100%0%in vitro (TK); in vivo (mouse, nasal inoculation)1 (100%)0%0%0%---[72]
2019, TurkeySertcelik Colistin23carbapenem-resistant (100%)100%26%in vitro (CB) 1 (4%)10 (43%)12 (52%)0%---[185]
2019, ChinaBian Colistin9carbapenem-resistant (100%)-0%in vitro (CB, TK) 1 (11%)------[186]
2018, ChinaZhuColistin21-100%61% (13/21)in vitro (CB) 0%2 (9%)19 (90%)0%--The authors reported 8 isolates to be colistin-R, but only 3 isolates had MIC > 2.[151]
2018, ThailandLeelasupasriColistin15carbapenem-resistant (100%)100%0%in vitro (CB, ET) 4 (26%)7 (46%)4 (26%)0%---[187]
2017, ThailandLertsrisatitColistin17CoR-AB; carbapenemase-producing; efflux-pump (100%)100%100%in vitro (CB, ET)---0%--Treatment in vivo (patients) with COL + FOS lead to death (2/2).[188]
2016, ChinaFanColistin12XDR (100%)100%0%in vivo (mouse, thigh-infection)model1 (8%)--0%---[189]
2016, Brazil Leite Colistin20OXA-23, OXA-143 (100%)100%35% (7/20)in vitro (CB, TK, 2-well) 0%------[83]
2015, ChinaWeiColistin50XDR (100%)94%50%in vitro (CB) 25 (50%)0%22 (44%)3 (6%) Synergism (FICI: =< 0.5). Indifference (FICI: 0.5–4). Antagonism (FICI: >= 4).[190]
2013, ChinaZhangPolymyxin B25PDR (100%)100%100%in vitro (TK) 4 (16%)11 (44%)10 (40%)0%0%25 (100%)-[65]
2011, ThailandSantimaleeworagunColistin8carbapenem-resistant (100%)0%-in vitro (CB, TK) 13%------[99]
N. gonorrhoeae2014, NetherlandsWind Colistin4---in vitro (ET)0%------[54]
Table 11. Studies on combination between fosfomycin and daptomycin. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 11. Studies on combination between fosfomycin and daptomycin. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainsYear and CountryAuthorNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Daptomycin-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%) Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Daptomycin Susceptibility Restoration (%)CommentsReference
S. aureus2019, TaiwanLee100MRSA (100%)15 (15%)0%in vitro (CB)37 (37%)44 (44%)19 (19%)0%--All isolates had MIC daptomycin = 1 (previously selected among 1353 isolates).[191]
2019, SpainCoronado-Alvarez4MRSA (50%)--in vitro (TK)4 (100%)0%0%0%--The authors also performed a retrospective review of 75 patients with severe Gram-positive infections and found that DAP + FOS (30) was the most effective combination.[63]
2018, SpainGarcìa-de-la-Mària5 (in vitro); 1 (in vivo)MRSA (100%)0%0%in vitro (TK), in vivo (rabbit, endocarditis)in vitro: 5 (100%); in vivo: 1 (100%)0%0%0%---[79]
2017, TurkeyAktas25MRSA (100%)11 (44%)0%in vitro (CB)25 (100%)0%0%0%---[80]
2015, AustriaLingscheid1MRSA (100%)0%0%in vivo (rats, implant-associated osteomyelitis)1 (100%)0%0%0%---[81]
2013, SpainGarrigós1MRSA (100%)0%0%in vitro (TK), in vivo (rat, foreign-body infection)in vitro: 0%; in vivo: 1 (100%)0%in vitro: 1 (100%)0%---[37]
2012, SpainMiró14MRSA (35%); GISA (14%)0%1 (7%)in vitro (TK)11 (79%)3 (21%)0%0%--The combination was bactericidal against 8 (57%) isolates. The authors also reported the case reports of 3 patients with S. aureus (1 MSSA, 2 MRSA) endocarditis successfully treated with high-dose DAP (10/kg/day) + FOS.[192]
2011, AustriaPoeppl1MRSA (100%)0%0%in vivo (rats, osteomyelitis)0%0%1 (100%)0%--FOS and FOS + DAP were significantly superior to placebo and to DAP alone. FOS + DAP was not more effective than FOS alone.[193]
E. faecalis2019, ChinaZheng4 (TK) + 4 (biofilm assay)-1 (12%)2 (25%)in vitro (TK, biofilm assay)TK: 4 (100%). Biofilm assay: 3 (75%)0%TK: 0%. Biofilm assay: 1 (25%)0%--TK performed on 4 linezolid-R isolates. Biofilm assay performed on 4 linezolid-S isolates. DAP + FOS demonstrated significantly more anti-biofilm activities then DAP or FOS alone.[194]
1992, USARice1-01 (100%)in vitro (TK), in vivo (rats, endocarditis)in vitro: 1 (100%)in vitro: 0%; in vivo: 1 (100%)0%0%-in vitro: 1 (100%)The isolate was highly R to gentamicin. DAP + FOS sterilized more valves (59% VS 35%) than DAP alone. Despite this, the combination in vivo was considered "additive" because it was not possible to demonstrate a statistically significant superiority in comparison with DAP alone.[82]
1989, USARice21-00in vitro (TK)21 (100%)0%0%0%--All isolates were highly R to gentamicin. The bactericidal effect of DAP alone was not increased by the addition of FOS.[195]
E. faecium2013, USADescourouez4VRE (100%)0%0%in vitro (TK)4 (100%)0%0%0% [196]--The combination resulted strongly bactericidal.[67]
Staphylococcus spp., Enterococcus spp. 1988, ItalyDebbia50-- in vitro (CB, TK)CB: 80%TK: 95%0%CB: 20%TK: 5%0%--A total of 50 strains was tested with CB, and only 20 strains were tested with TK.[197]
Table 12. Studies on combination between fosfomycin and tigecycline. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 12. Studies on combination between fosfomycin and tigecycline. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Tigecycline-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%) Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Tigecycline Susceptibility Restoration (%)CommentsReference
Enterobacterales2019, USAFlamm207/30 MDR strains (A. baumannii, Enterobacterales e P. aeruginosa) included 2 ESBL e 2 KPC Enterobacterales--in vitro (CB)5 (25%)10 (50%)5 (25%)0%--Authors considered Partial Sinergy when FICI was between 0.5–1 and Additive effect for FICI = 1. [38]
2017, TaiwanKu9ESBL KP producing (100%)4 (44,4%)4 (44%)in vitro (TK)6 (66%)0%3 (33%)0%---[84]
2011, FranceBerçot9NDM-1 KPC (100%)2 (22%)3 (33%)in vitro (CB)0%-9 (100%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4.[85]
E. coli2013, SwitzerlandCorvec1Bj HDE-1 (100%) (ESBL and Ciprofloxacin resistant)0%0%in vitro (TK); in vivo (Guinea pigs, cage infection)TK: 0%; in vivo: 0%TK: 100%; in vivo: 0%TK: 0%; in vivo: 100%0%---[73]
2011, GreeceSamonis20ESBL (100%)0%1 (5%)in vitro (ET)5 (25%)-15 (75%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4. In vivo experiment: bacterial count using FOS + TIG combination was reduced ≥ 2 log over single antimicrobials[86]
K. pneumoniae2019, ChinaHuang30KPC (100%)19 (63%)11 (36%)in vitro (ET, CB)ET: 5 (16%); CK: 4 (13%)ET: 9 (30%); CK: 11 (36%)ETt: 16 (53%); CK: 15 (50%)0%ET: 14/19 (73%); CK: 6/15 (40%)ET: 5/11 (45%); CK: 7/13 (53,%)ET and CB showed different rates of FOS and TIG resistance and different rates of susceptibility restoration; otherwise the 2 methods had similar resulted in establishing synergistic, additive or indifferent effect.[88]
2019, GreecePapoutsaki11KPC (100%)35%96%in vitro (ET, TK)ET: 16/33 (48%); TKA: 1/22 (4%)ET: 17/33 (51%); TKA: 21/22 (95%)0%0%--ET was performed three times with different methods: a) Etest/Agar method; b) Cross formation method; c) MIC/MIC ratio method. TK was performed two times: a) TIG 1,3 mg/L + FOS 0,5xMIC and b) TIG 1,3 mg/L + FOS 30 mg/L.[87]
2017, ChinaYu136KPC (100%)78 (57%)25 (18%)in vitro (CB, TK)CK: 2 (1%); TKA: 0%CK: 113 (83%); TKA: 3 (75%)CK: 19 (14%); TKA: 1 (25%)CK: 2 (1%); TKA: 0%--Only 4 strains were tested with TK.[89]
2013, TurkeyEvren12OXA-48 (100%)11 (92%)5 (41%)in vitro (CB)4 (33%)6 (50%)2 (16%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4. In vivo experiment: bacterial count using FOS + TIG combination was reduced ≥ 2 log over single antimicrobials[74]
2011, GreeceSamonis65Serine-KPC (77%) - MBL (1%) - ESBL (21%)1 (1%)10 (15%)in vitro (ET)18 (27%)-47 (72%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4. In vivo experiment: bacterial count using FOS + TIG combination was reduced ≥ 2 log over single antimicrobials[86]
P. aeruginosa2011, GreeceSamonis15MDR (100%)1 (6%)15 (100%)in vitro (ET)2 (13%)-13 (86%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4. In vivo experiment: bacterial count using FOS + TIG combination was reduced ≥ 2 log over single antimicrobials[86]
A. baumannii2019, USAFlamm57/30 MDR strains (A. baumannii, Enterobacterales e P. aeruginosa)--in vitro (CB)0%4 (80%)1 (20%)0%--Authors considered Partial Sinergy when FICI was between 0.5–1 and Additive effect for FICI = 1. [38]
2016, NetherlandsLeite20Colistin-Resistant (65%)20 (100%)5%in vitro (CB, 2-Well Method)0%-----Any synergistic effect was reported. Additive, Indifferent and antagonistic effect were not evaluated.[83]
S. aureus2018, ItalySimonetti15MRSA (100%)00%in vitro (CB); in vivo (mice, wound infection)12 (80%)-3 (20%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4. In vivo experiment: bacterial count using FOS + TIG combination was reduced ≥ 2 log over single antimicrobials.[90]
2012, TaiwanTang33 (8 TK)MRSA (100%)6%0%in vitro (TK, Biofilm MTT-staining method)0%-100%0%--Only 8 strains were tested with Time–kill Assay. Biofilm cultures were 100% TIG resistant and 94% FOS resistant. No FICI were reported by authors, no synergistic effect was seen on any strains.[69]
E. faecalis2018, ItalySimonetti15-0%0%in vitro (CB); in vivo (mice, wound infection)12 (80%)-3 (20%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4. In vivo experiment: bacterial count using FOS + TIG combination was reduced ≥ 2 log over single antimicrobials.[90]
2013, TaiwanTang9VRE (100%)56%0%in vitro (TK, Biofilm Model)TKA: 3 (33%); BM: 5 (56%)-----Additive, Indifferent and antagonistic effect were not evaluated.[13]
E. faecium2019, ThailandHemapampairoa12VRE (100%)12 (100%)3 (25%)in vitro (CB)1 (8%)9 (75%)2 (16)%0%---[55]
2018, ItalySimonetti15-0%0%in vitro (CB)10 (66)%-5 (33%)0%--Authors considered Indifferent effect for FICI between 0.5 and 4.[90]
2013, TaiwanTang10VRE (100%)70%0%in vitro (TK, Biofilm Model)TKA: 3 (30%); BM: 1 (10%)-----Additive, Indifferent and antagonistic effect were not evaluated.[13]
N. gonorrhoeae2015, NetherlandsWind4Azithromycin and Ceftriaxone Resistant (100%)--in vitro (ET)0%0%4 (100%)----[54]
Table 13. Studies on combination between fosfomycin and linezolid. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 13. Studies on combination between fosfomycin and linezolid. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Linezolid-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Linezolid Susceptibility Restoration (%)CommentsReference
S. aureus2018, ChinaChen11 (3 TK)MRSA (50%)0%0%in vitro (CB, TK)CK: 8 (72%); TK: 3 (100%)CK: 3 (27%); TK: 0%CK: 0%; TK: 0%CK: 0%; TK: 0%--Only 3 strains were tested with TK. For the same 3 strains, the authors also evaluated. Post-Antibiotic Effect (PAE) of LZD alone and in combination with FOS. PAE of LZD + FOS seemed to be increased with the increase in time of exposure, even if no statistically significant difference was found.[198]
2018, SpainCoronado-Alvarez2MRSA (100%)--in vitro (TK)2 (100%)0%0%0%--Synergy was defined as a reduction > 3 log CFU/mL over antimicrobial agent alone, additive effect was defined as areduction < 3 log CFU/mL. Synergistic effect was demonstrated only when 4 x MIC LZD + 2 x MIC FOS were used; 1 × MIC LZD + 2 × MIF FOS regimen showed Additive effect.[63]
2016, ChinaChai3 (1 TK)MRSA (100%)2 (66%)0%in vitro (CB, TK)CK: 3 (100%); TK: 1 (100%)CK: 0%; TK: 0%CK: 0%; TK: 0%CK: 0%; TK: 0%--Only 1 strain was tested with Time–kill Assay. The authors also evaluated in vitro and in vivo efficacy of LIN + FOS on MRSA biofilm (all 3 strains), demonstrating a synergistic effect only in vitro when using 1/2 MIC LZD + 1/2 MIC FOS and not with lower concentrations.[94]
2014, ChinaXu-Hong102MRSA (100%)**MIC range 16-128 mg/L0%in vitro (CB)100 (98%)-2 (2%)0%100%100%The authors considered Indifferent effect for FICI between 0.5 and 4. Fosfomycin MIC range in combination was 2-32 mg/L, LZD MIC in combination was 0,125–1 mg/L.[199]
2012, TaiwanTang33 (8 TK)MRSA (100%)6%0%in vitro (TK, Biofilm MTT-staining method)------Only 8 strains were tested with Time–kill Assay. Biofilm cultures were 100% LZD resistant and 94% FOS resistant. Combination of FOS + LZD determined a statistically significant reduction on ODRs in biofilm cultures.[69]
2010, SpainPachón-Ibáñez1GISA 100% (Gentamicin Intermediate S. aureus)--in vitro (TK); in vivo (Murine peritonitis model)1 (100%)0%0%0%--In vivo experiment on mice showed a higher rate of blood culture negativization when using FOS + LZD therapy (57%) then using FOS or LZD alone (43% and 27% respectively).[36]
2006, SpainSahuquillo Arce5 (4 TK)-0%0%in vitro (CB, TK)CK: 4 (80%); TK: 4 (100%)CK: 1 (20%); TK: 0%CK: 0%; TK: 0%CK: 0%; TK: 0%--Synergistic effect at CB was confirmed with TK on 4 strains.[200]
2001, AustriaGrif5 (1 TK)MRSA (60%)0%0%in vitro (CB, TK, TEM)CK: 5 (100%); TK: 0%-CK: 0%; TK: (1) 100%CK: 0%; TK: 0%--The authors did not consider additive effect. They also performed Transmission Electron Microscopy, demonstrating profound morphological alteration of 2 strains when using FOS + LZD, which were not seen using FOS or LZD alone.[43]
2018, ChinaLi4MRSA (50%)0%0%in vitro (CB, TK); in vivo (Galleria melonella Survival Assay)CK: 4 (100%); TK: 4 (100%)CK: 0%; TK: 0%CK: 0%; TK: 0%CK: 0%; TK: 0%--TKA showed synergism, but bacteriostatic effect. In vivo experiment showed statistically significant higher efficacy of high-dose LZD + FOS combination, then high dose of FOS or LZD alone, but low-dose combination had no significant differences with monotherapy orhigh-dose combination.[95]
S. epidermidis2001, AustriaGrif2-0%0%in vitro (CB)2 (100%)-0%0%--The authors did not consider additive effect. They also performed Transmission Electron Microscopy, demonstrating profound morphological alteration of 2 strains when using FOS + LZD, which were not seen using FOS or LZD alone.[43]
E. faecalis2013, TaiwanTang9VRE (100%)56%0%in vitro (TK, Biofilm Model)TKA: 0%; BM: 0%-----The authors did not consider additive, indifferent or antagonistic effect.[13]
2019, ChinaQi2VRE (50%)2 (100%)0%in vitro (CB, TK, TEM)CK: 0%; TK: 0%)CK: 2 (100%); TK: 1 (50%)CK: 0%; TK: 1 (50%)CK: 0%; TK: 0%2 (100%)2 (100%)Transmission Electron Microscopy, demonstrated more morphological alterations when using FOS + LZD, then using FOS or LZD alone.[201]
E. faecium2019, ThailandHemapampairoa12VRE (100%)12 (100%)0%in vitro (CB)3 (25%)9 (75%)0%0%---[55]
2013, TaiwanTang10VRE (100%)70%80%in vitro (TK, Biofilm Model)TKA: 1 (10%); BM: 0%-----The authors did not consider additive, indifferent or antagonistic effect.[13]
2012, USADescourouez32VRE (100%)9%3%in vitro (TK)See commentsSee comments0%0%--The authors considered MIC ≤ 64 mg/L as FOS breakpoint. FOS combined with LZD was either synergistic or additive yet bacteriostatic. Percentages of strains on which there was synergistic effect were not reported [67]
2019, ChinaQi4VRE (75%)4 (100%)1 (25%)in vitro (CB, TK, TEM); in vivo (Galleria Melonella Survival Assay)CK: 2 (50%); TK: 2 (50%)CK: 1 (25%); TK: 1 (25%)CK: 1 (25%); TK: 1 (25%)0%3 (75%)4 (100%)Transmission Electron Microscopy, demonstrated more morphological alterations when using FOS + LZD, then using FOS or LZD alone. In vivo experiment showed higher survival rates of larvae when using FOS + LZD then LZD alone, but similar rates using FOS alone.[201]
Table 14. Studies on combination between fosfomycin and rifampin. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
Table 14. Studies on combination between fosfomycin and rifampin. CB: checkerboard assay; TK: time–kill assay; ET: E-test.
StrainYear and CountryAuthorNumber of IsolatesKnown Resistance Mechanisms or Determinants (%)FOS-Resistant (%)Rifampin-Resistant (%)In Vitro (Methods)/In Vivo (Animal and Site of Infection)Synergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)FOS Susceptibility Restoration (%)Rifampin Susceptibility Restoration (%)CommentsReference
E. coli1978, SpainOlay17---in vitro (CB); in vivo (mouse, peritonitis)1 (5,9%)9 (52,9%)7 (41,2%)0%---[14]
A. baumannii2016, BrazilLeite20OXA-51, OXA-23, OXA-143 (100%)20 (100%)20 (100%)in vitro (CB, TK)0%------[83]
S. aureus2018, ItalySimonetti16MRSA (100%)0%2 (12%)in vitro (CB, TK); in vivo (mouse, wound infection)16 (100%)0%0%0%---[90]
2014, SwitzerlandMihailescu1MRSA (100%)0%0%in vitro (ET, TK); in vivo (foreign-body infection model)in vitro: 1 (100%); in vivo: 100% at day 120%0%0%---[96]
2013, ChinaTang8MRSA (100%)0%8 (100%)in vitro (biofilm assay)4 (50%)------[91]
2012, SpainGarrigos1MRSA (100%)--in vitro (TK); in vivo (rat, tissue cage infection)in vivo: 1 (100%) at day 8 and day 11--in vitro: 1 (100%)--In vitro FOS antagonized the effect of RIF.[37]
2012, TaiwanTang33MRSA (100%)6% (planktonic) 94% (biofilm)0% (planktonic) 79% (biofilm)in vitro (TK)0%------[69]
2001, AustriaGrif5MRSA (100%)--in vitro (CB, TK)100%------[43]
1987, FranceQuentin6-33%0%in vitro (TK)0%0%33%33%--RIF antagonizes FOS. In particular, it antagonizes FOS against susceptible and intermediate isolates to RIF. The combination resulted indifferent against RIF-resistant isolates. For 2 isolates it was not possible to infer their susceptibility to RIF.[35]
1984, GermanyTraub6GRMR (100%)0%0%in vitro (CB); in vivo (mouse, peritonitis)--2 (33%)----[202]
1978, SpainOlay38---in vitro (CB); in vivo (mouse, peritonitis)13 (34%)24 (63%)1 (2%)----[14]
S. pneumoniae1994, FranceDoit26-0%0%in vitro (TK)0%0%100%0%---[134]
S. agalactiae, S. pyogenes, S. oralis2017, GermanyGonzalez Moreno3-33%0%in vitro (ET)1 (100%) S. oralis-1 (100%) S. agalactiae; 1 (100%) S. pyogenes----[9]
E. feacalis2018, ItalySimonetti16-0%2 (12%)in vitro (CB, TK); in vivo (mouse, wound infection)12 (75%)0%4 (25%)*0%--*The FICIs were interpreted as indifferent if > 0.5 and < 4.[90]
2013, TaiwanTang9VRE (100%)56%11%in vitro (TK, biofilm)TK: 3 (33%); biofilm: 9 (100%)--0%---[13]
E. faecium2018, ItalySimonetti15-0%2 (13%)in vitro (CB, TK); in vivo (mouse, wound infection)11 (73%)0%4 (27%)*0%--*The FICIs were interpreted as indifferent if > 0.5 and < 4.[90]
2013, TaiwanTang10VRE (100%)70%90%in vitro (TK)TK: 2 (20%); biofilm: 4 (40%)------[13]
S. epidermidis2011, AustriaGrif2MRSA (100%)--in vitro (CB, TK)2 (100%)------[43]
1987, FranceQuentin3-NANAin vitro (TK)0%0%50%---For 1 isolate it was not possible to infer its susceptibility to RIF.[35]
N. gonorrhoeae2015, NetherlandsWind4---in vitro (ET)1 (25%)------[54]
Table 15. Effect of FOS in combination with different antibiotics: overview.
Table 15. Effect of FOS in combination with different antibiotics: overview.
Antibiotic ClassStrainsNumber of StudiesNumber of IsolatesSynergistic Effect (%)Additive Effect (%)Indifferent Effect (%)Antagonistic Effect (%)Comments
Penicillins, penicillins + β-lactamase inhibitors, penicillinase-resistant penicillinsEnterobacterales9267511928 One study [11] reported high rates of indifferent effect of FOS + PIP/TAZ against PIP/TAZ-R isolates.
P. aeruginosa6235154045 -
Acinetobacter spp.1560200 -
Staphylococcus spp. 7295421533 -
Streptococcus spp.6119305515 -
Enterococcus spp.4602504210Antagonistic effect observed in biofilms of some E. faecalis isolates.
Cephalosporins, cephalosporins + β-lactamase inhibitorsEnterobacterales8251333320 One study [11] reported high rates of indifferent effect of FOS + 4 different cephalosporins against cephalosporin-R isolates.
P. aeruginosa133183640231Antagonistic effect against 4 P. aeruginosa isolates [22].
Acinetobacter spp.239833 Effect of the combination indeterminate on 33 isolates.
Staphylococcus spp.12284571291Great heterogeneity of results.
Streptococcus spp.66333598 -
Enterococcus spp.27778022 -
N. gonorrhoeae3440595 -
CarbapenemsEnterobacterales23542433719
P. aeruginosa154452925361-
Acinetobacter spp.5103281722 -
Gram + cocci122315613228S. aureus, S. epidermidis, Enterococci spp., S. pneumoniae. High rates of antagonistic effect reported on E. faecalis isolates.
N. gonorrhoeae1407525 -
MonobactamsEnterobacterales471152745 -
P. aeruginosa3138295417 -
QuinolonesEnterobacterales6264171269 -
P. aeruginosa182634236385Synergism rates not concordant in all studies.
Acinetobacter spp.3412107 -
Staphylococcus spp.79037934 -
N. gonorrhoeae1400100 -
AminoglycosidesEnterobacterales19713203136 Synergism rates not concordant in all studies.
P. aeruginosa234404329271Synergism rates not concordant in all studies.
Acinetobacter spp.510237518 Synergism rates not concordant in all studies.
S. aureus8301264531Antagonistic effect of FOS + gentamicin against 4 isolates [12].
Streptococcus spp.11605248 -
E. faecium18621325 -
N. gonorrhoeae1402575 -
H. influenzae1100100 -
GlycopeptidesA. baumannii12000100 -
Staphylococcus spp.122291716652In 2 studies [69,176] VAN exhibited higher synergistic rates than TEC. Antagonistic effect with FOS + VAN against 5 isolates of S. aureus [12,43].
Enterococcus spp.255552224 -
S. pneumoniae2284492 -
MacrolidesEnterobacterales187533414 -
N. gonorrhoeae21200100 -
P. aeruginosa23119792 -
S. aureus13426686 -
S. epidermidis11100100 -
S. pseudointermedius18622512 -
Streptococcus spp.126152758 Only erythromycin was tested in combination with FOS. Against almost half of strains additive or, less frequently, synergistic effect was observed.
TetracyclinesEnterobacterales212051184 Indifferent effect when tetracycline was tested, but one study showed additive or synergistic effect when using minocycline + FOS combination [38].
P. aeruginosa1540600 -
Acinetobacter spp.125125632 In all experiment minocycline susceptibility restoration was observed [65].
S. aureus315272127 -
Enterococcus spp.375241020 Indifferent effect when minocycline was tested, but one study showed additive or synergistic effect when using doxicycline + FOS combination [68].
N. gonorrhoeae1400100 -
PolymyxinsEnterobacterales183812635354Antagonistic effect of FOS + colistin observed against 14 isolates of K. pneumoniae.
P. aeruginosa4111274131 -
Acinetobacter spp.122061915321Antagonistic effect of FOS + colistin observed against 3 isolates of A. baumannii.
N. gonorrhoeae1400100 -
DaptomycinStaphylococcus spp.13186563114 -
Enterococcus spp.5499703 -
TigecyclineEnterobacterales93131744341One in vivo study observed indifferent effect in 100% of cases against E. coli [73] and one in vitro study reported 2 cases of antagonistic effect against K. pneumoniae isolates [89].
P. aeruginosa11513087 -
Acinetobacter spp.2250164 -
S. aureus24821079 Conflicting results (total indifference or almost total synergistic effect).
Enterococcus spp.3616109 -
N. gonorrhoeae1400100 -
LinezolidEnterococcus spp.46917296 Synergistic effect was never observed for E. faecalis (2 studies) [13,201].
S. aureus91667422 -
S. epidermidis1210000 -
RifampinE. coli11765341 -
A. baumannii12000100 -
S. aureus9114352143Antagonistic effect of FOS + RIF against 3 isolates [35,37].
S. epidermidis2540040 -
Streptococcus spp.2293097 -
Enterococcus spp.25059012 -
N. gonorrhoeae1425075 -
MetronidazoleIntestinal bacteria (not specified)1NA--- -
H. pylori12402180 -
SpectinomycinN. gonorrhoeae1400100 -
SulbactamA. baumannii1875025 -
LincomycinS. aureus13781190 -
NitroxolineP. aeruginosa1812088 -
Dalfopristin-QuinupristinStaphylococcus spp.21210000 -
Fusidic acidS. aureus323963433 -
ChloramphenicolEnterobacterales4468393425 -
P. aeruginosa119533710 -
S. aureus148443719 -
NitrofurantoinEnterobacterales110000100 -
Enterococcus spp.13200100 -
Trimethoprim-SulfamethoxazoleEnterobacterales21202589 -
S. aureus114830953Antagonistic effect was reported for 4 isolates [12].

Supplementary Materials

The following are available online at https://www.mdpi.com/2079-6382/9/8/500/s1, Table S1: Studies on combination between fosfomycin and different antibiotics. CB: checkerboard assay; TK: time–kill assay; ET: E-test, Table S2: Studies on combination between fosfomycin and molecules other than antibiotics. CB: checkerboard assay; TK: time–kill assay.

Author Contributions

Conceptualization, R.M.A., R.L. and S.D.B.; methodology, A.E.M.; investigation, R.M.A., L.P., V.V., R.P., M.F., F.M., A.L., S.D.B.; data curation, R.M.A., A.E.M.; writing-original draft preparation, R.M.A., L.P., M.F, F.M., R.L. and S.D.B.; writing-review and editing, R.L. and S.D.B.; supervision, R.L. and S.D.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The Authors are grateful to Maria Crapulli for her precious help in finding articles available only in paper version.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Falagas, M.E.; Vouloumanou, E.K.; Samonis, G.; Vardakas, K.Z. Fosfomycin. Clin. Microbiol. Rev. 2016, 29, 321–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hendlin, D.; Stapley, E.O.; Jackson, M.; Wallick, H.; Miller, A.K.; Wolf, F.J.; Miller, T.W.; Chaiet, L.; Kahan, F.M.; Foltz, E.L.; et al. Phosphonomycin, a new antibiotic produced by strains of streptomyces. Science 1969, 166, 122–123. [Google Scholar] [CrossRef] [PubMed]
  3. 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] [PubMed] [Green Version]
  4. Falagas, M.E.; Kastoris, A.C.; Kapaskelis, A.M.; Karageorgopoulos, D.E. Fosfomycin for the treatment of multidrug-resistant, including extended-spectrum beta-lactamase producing, Enterobacteriaceae infections: A systematic review. Lancet. Infect. Dis. 2010, 10, 43–50. [Google Scholar] [CrossRef]
  5. Zhanel, G.G.; Zhanel, M.A.; Karlowsky, J.A. Oral Fosfomycin for the Treatment of Acute and Chronic Bacterial Prostatitis Caused by Multidrug-Resistant Escherichia coli. Can. J. Infect. Dis. Med. Microbiol. = J. Can. des Mal. Infect. la Microbiol. Medicale 2018, 2018, 1404813. [Google Scholar] [CrossRef] [Green Version]
  6. Petrosillo, N.; Taglietti, F.; Granata, G. Treatment Options for Colistin Resistant Klebsiella pneumoniae: Present and Future. J. Clin. Med. 2019, 8, 934. [Google Scholar] [CrossRef] [Green Version]
  7. Krcmery, S.; Hromec, J.; Demesova, D. Treatment of lower urinary tract infection in pregnancy. Int. J. Antimicrob. Agents 2001, 17, 279–282. [Google Scholar] [CrossRef]
  8. Behera, B.; Mohanty, S.; Sahu, S.; Praharaj, A. In vitro activity of fosfomycin against multidrug-resistant urinary and nonurinary Gram-negative isolates. Indian, J. Crit. Care Med. 2018, 22, 533–536. [Google Scholar] [CrossRef]
  9. Gonzalez Moreno, M.; Trampuz, A.; Di Luca, M. Synergistic antibiotic activity against planktonic and biofilm-embedded Streptococcus agalactiae, Streptococcus pyogenes and Streptococcus oralis. J. Antimicrob. Chemother. 2017, 72, 3085–3092. [Google Scholar] [CrossRef]
  10. EUCAST: Clinical breakpoints and dosing of antibiotics. Available online: https://eucast.org/clinical_breakpoints/ (accessed on 12 April 2020).
  11. Avery, L.M.; Sutherland, C.A.; Nicolau, D.P. In vitro investigation of synergy among fosfomycin and parenteral antimicrobials against carbapenemase-producing Enterobacteriaceae. Diagn. Microbiol. Infect. Dis. 2019, 95, 216–220. [Google Scholar] [CrossRef]
  12. Alvarez, S.; Jones, M.; Berk, S.L. In vitro activity of fosfomycin, alone and in combination, against methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 1985, 28, 689–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Tang, H.-J.; Chen, C.-C.; Zhang, C.-C.; Su, B.-A.; Li, C.-M.; Weng, T.-C.; Chiang, S.-R.; Ko, W.-C.; Chuang, Y.-C. In vitro efficacy of fosfomycin-based combinations against clinical vancomycin-resistant Enterococcus isolates. Diagn. Microbiol. Infect. Dis. 2013, 77, 254–257. [Google Scholar] [CrossRef] [PubMed]
  14. Olay, T.; Rodriguez, A.; Oliver, L.E.; Vicente, M.V.; Quecedo, M.C.R. Interaction of fosfomycin with other antimicrobial agents: In vitro and in vivo studies. J. Antimicrob. Chemother. 1978, 4, 569–576. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Chavanet, P.; Peyrard, N.; Pechinot, A.; Buisson, M.; Duong, M.; Neuwirth, C.; Kazmierczak, A.; Portier, H. In vivo activity and pharmacodynamics of amoxicillin in combination with fosfomycin in fibrin clots infected with highly penicillin-resistant Streptococcus pneumoniae. Antimicrob. Agents Chemother. 1996, 40, 2062–2066. [Google Scholar] [CrossRef] [Green Version]
  17. Vicente, M.V.; Olay, T.; Rodriguez, A. Experimental endocarditis caused by Streptococcus sanguis: Single and combined antibiotic therapy. Antimicrob. Agents Chemother. 1981, 20, 10–14. [Google Scholar] [CrossRef] [Green Version]
  18. Nakazawa, H.; Kikuchi, Y.; Honda, T.; Isago, T.; Nozaki, M. Enhancement of antimicrobial effects of various antibiotics against methicillin-resistant Staphylococcus aureus (MRSA) by combination with fosfomycin. J. Infect. Chemother. 2003, 9, 304–309. [Google Scholar] [CrossRef]
  19. Courcol, R.J.; Martin, G.R. In-vitro activity of the combination of ceftriaxone and fosfomycin against staphylococci. J. Antimicrob. Chemother. 1987, 19, 276–278. [Google Scholar] [CrossRef]
  20. Portier, H.; Lucht, F.; Chavanet, P.; Kazmierczak, A.; Tremeaux, J.C.; Duez, J.M. Cefotaxime in combination with other antibiotics for the treatment of severe methicillin-resistant staphylococcal infections. Infection 1985, 13, S123–S128. [Google Scholar] [CrossRef]
  21. 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, e00779-19. [Google Scholar] [CrossRef] [Green Version]
  22. Pruekprasert, P.; Tunyapanit, W. In vitro activity of fosfomycin-gentamicin, fosfomycin-ceftazidime, fosfomycin-imipenem and ceftazidime-gentamicin combinations against ceftazidime-resistant Pseudomonas aeruginosa. Southeast Asian, J. Trop. Med. Public Health 2005, 36, 1239–1242. [Google Scholar]
  23. Chavanet, P.; Beloeil, H.; Pechinot, A.; Duigou, F.; Buisson, J.C.; Duong, M.; Neuwirth, C.; Kazmierczak, A.; Portier, H. In vivo activity and pharmacodynamics of cefotaxime or ceftriaxone in combination with fosfomycin in fibrin clots infected with highly penicillin- resistant Streptococcus pneumoniae. Antimicrob. Agents Chemother. 1995, 39, 1736–1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ribes, S.; Taberner, F.; Domenech, A.; Cabellos, C.; Tubau, F.; Liñ Ares, J.; Ferná Ndez Viladrich, P.; Gudiol, F. Evaluation of fosfomycin alone and in combination with ceftriaxone or vancomycin in an experimental model of meningitis caused by two strains of cephalosporin-resistant Streptococcus pneumoniae. J. Antimicrob. Chemother. 2006, 57, 931–936. [Google Scholar] [CrossRef]
  25. Utsui, Y.; Ohya, S.; Magaribuchi, T.; Tajima, M.; Yokota, T. Antibacterial activity of cefmetazole alone and in combination with fosfomycin against methicillin- and cephem-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 1986, 30, 917–922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kazmierczak, A.; Pechinot, A.; Duez, J.M.; Kohli, E.; Tremeaux, J.C.; Portier, H. Bactericidal activity of cefotaxime and fosfomycin in cerebrospinal fluid during the treatment of rabbit meningitis experimentally induced by methicillin-resistant staphylococcus aureus. Infection 1985, 13, S76–S80. [Google Scholar] [CrossRef]
  27. Chavanet, P.; Muggeo, E.; Waldner, A.; Dijoux, S.; Caillot, D.; Portier, H. Synergism between cefotaxime and fosfomycin in the therapy of methicillin and gentamicin resistant Staphylococcus aureus infection in rabbits. Eur. J. Clin. Microbiol. Infect. Dis. 1990, 9, 271–275. [Google Scholar] [CrossRef]
  28. Del Río, A.; García-de-la-Mària, C.; Entenza, J.M.; Gasch, O.; Armero, Y.; Soy, D.; Mestres, C.A.; Pericás, J.M.; Falces, C.; Ninot, S.; et al. Fosfomycin plus β-Lactams as Synergistic Bactericidal Combinations for Experimental Endocarditis Due to Methicillin-Resistant and Glycopeptide-Intermediate Staphylococcus aureus. Antimicrob. Agents Chemother. 2016, 60, 478–486. [Google Scholar] [CrossRef] [Green Version]
  29. Papp-Wallace, K.M.; Zeiser, E.T.; Becka, S.A.; Park, S.; Wilson, B.M.; Winkler, M.L.; D’Souza, R.; Singh, I.; Sutton, G.; Fouts, D.E.; et al. Ceftazidime-Avibactam in Combination With Fosfomycin: A Novel Therapeutic Strategy Against Multidrug-Resistant Pseudomonas aeruginosa. J. Infect. Dis. 2019, 220, 666–676. [Google Scholar] [CrossRef] [Green Version]
  30. Lefort, A.; Chau, F.; Lepeule, R.; Dubée, V.; Kitzis, M.D.; Dion, S.; Fantin, B. Activity of fosfomycin alone or combined with cefoxitin in vitro and in vivo in a murine model of urinary tract infection due to Escherichia coli harbouring CTX-M-15-type extended-spectrum β-lactamase. Int. J. Antimicrob. Agents 2014, 43, 366–369. [Google Scholar] [CrossRef]
  31. Ojdana, D.; Gutowska, A.; Sacha, P.; Majewski, P.; Wieczorek, P.; Tryniszewska, E. Activity of Ceftazidime-Avibactam Alone and in Combination with Ertapenem, Fosfomycin, and Tigecycline Against Carbapenemase-Producing Klebsiella pneumoniae. Microb. Drug Resist. 2019, 25, 1357–1364. [Google Scholar] [CrossRef]
  32. Cuba, G.; Rocha-Santos, G.; Cayô, R.; Streling, A.; Nodari, C.; Gales, A.; Pignatari, A.; Nicolau, D.; Kiffer, C. In Vitro Synergy of Ceftolozane/Tazobactam in Combination With Fosfomycin or Aztreonam Against MDR Pseudomonas Aeruginosa. J. Antimicrob. Chemother. 2020, 75, 1874–1878. [Google Scholar] [CrossRef] [PubMed]
  33. Avery, L.M.; Sutherland, C.A.; Nicolau, D.P. Prevalence of in vitro synergistic antibiotic interaction between fosfomycin and nonsusceptible antimicrobials in carbapenem-resistant Pseudomonas aeruginosa. J. Med. Microbiol. 2019, 68, 893–897. [Google Scholar] [CrossRef] [PubMed]
  34. Monogue, M.; Nicolau, D. Antibacterial Activity of Ceftolozane/Tazobactam Alone and in Combination With Other Antimicrobial Agents Against MDR Pseudomonas Aeruginosa. J. Antimicrob. Chemother. 2018, 73, 942–952. [Google Scholar] [CrossRef] [PubMed]
  35. Quentin, C.; Saivin, S.; Lafferriere, C.; Noury, P.; Bebear, C. In vitro activity of fosfomycin combined with rifampin, pefloxacin and imipenem against staphylococci: A study by the time–kill curve method. Drugs Exp. Clin. Res. 1987, 13, 219–224. [Google Scholar] [PubMed]
  36. Pachón-Ibáñez, M.E.; Ribes, S.; Domínguez, M.Á.; Fernández, R.; Tubau, F.; Ariza, J.; Gudiol, F.; Cabellos, C. Efficacy of fosfomycin and its combination with linezolid, vancomycin and imipenem in an experimental peritonitis model caused by a Staphylococcus aureus strain with reduced susceptibility to vancomycin. Eur. J. Clin. Microbiol. Infect. Dis. 2011, 30, 89–95. [Google Scholar] [CrossRef] [PubMed]
  37. Garrigós, C.; Murillo, O.; Lora-Tamayo, J.; Verdaguer, R.; Tubau, F.; Cabellos, C.; Cabo, J.; Ariza, J. Fosfomycin-daptomycin and other fosfomycin combinations as alternative therapies in experimental foreign-body infection by methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2013, 57, 606–610. [Google Scholar] [CrossRef] [Green Version]
  38. Flamm, R.K.; Rhomberg, P.R.; Lindley, J.M.; Sweeney, K.; Ellis-Grosse, E.J.; Shortridge, D. Evaluation of the Bactericidal Activity of Fosfomycin in Combination with Selected Antimicrobial Comparison Agents Tested against Gram-Negative Bacterial Strains by Using Time–kill Curves. Antimicrob. Agents Chemother. 2019, 63, e02549-18. [Google Scholar] [CrossRef] [Green Version]
  39. Okazaki, M.; Suzuki, K.; Asano, N.; Araki, K.; Shukuya, N.; Egami, T.; Higurashi, Y.; Morita, K.; Uchimura, H.; Watanabe, T. Effectiveness of fosfomycin combined with other antimicrobial agents against multidrug-resistant Pseudomonas aeruginosa isolates using the efficacy time index assay. J. Infect. Chemother. 2002, 8, 37–42. [Google Scholar] [CrossRef]
  40. Mikuniya, T.; Kato, Y.; Ida, T.; Maebashi, K.; Monden, K.; Kariyama, R.; Kumon, H. Treatment of Pseudomonas aeruginosa biofilms with a combination of fluoroquinolones and fosfomycin in a rat urinary tract infection model. J. Infect. Chemother. 2007, 13, 285–290. [Google Scholar] [CrossRef]
  41. Bugnon, D.; Potel, G.; Xiong, Y.Q.; Caillon, J.; Navas, D.; Gras, C.; Kergueris, M.F.; Le Conte, P.; Jehl, F.; Baron, D.; et al. Bactericidal effect of pefloxacin and fosfomycin against Pseudomonas aeruginosa in a rabbit endocarditis model with pharmacokinetics of pefloxacin in humans simulated in vivo. Eur. J. Clin. Microbiol. Infect. Dis. 1997, 16, 575–580. [Google Scholar] [CrossRef]
  42. El-Wafa, W.M.A.; Ibrahim, Y.M. In Vitro Activity of Fosfomycin in Double and Triple Combinations with Imipenem, Ciprofloxacin and Tobramycin Against Multidrug-Resistant Escherichia coli. Curr. Microbiol. 2020, 77, 1–7. [Google Scholar] [CrossRef] [PubMed]
  43. Grif, K.; Dierich, M.; Pfaller, K.; Miglioli, P.; Allerberger, F. In vitro activity of fosfomycin in combination with various antistaphylococcal substances. J. Antimicrob. Chemother. 2001, 48, 209–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Thauvin, C.; Lemeland, J.F.; Humbert, G.; Fillastre, J.P. Efficacy of pefloxacin-fosfomycin in experimental endocarditis caused by methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 1988, 32, 919–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ullmann, U. Synergism between ciprofloxacin and fosfomycin in vitro. Infection 1987, 15, 264. [Google Scholar] [CrossRef]
  46. Mikuniya, T.; Kato, Y.; Kariyama, R.; Monden, K.; Hikida, M.; Kumon, H. Synergistic effect of fosfomycin and fluoroquinolones against Pseudomonas aeruginosa growing in a biofilm. Acta Med. Okayama 2005, 59, 209–216. [Google Scholar] [CrossRef]
  47. Krause, K.M.; Serio, A.W.; Kane, T.R.; Connolly, L.E. Aminoglycosides: An overview. Cold Spring Harb. Perspect. Med. 2016, 6, a027029. [Google Scholar] [CrossRef] [Green Version]
  48. Dos Santos Llma, D.A.F.; Do Nascimento, M.M.P.; Vitali, L.H.; Martinez, R. In vitro activity of antimicrobial combinations against multidrug-resistant Pseudomonas aeruginosa. Rev. Soc. Bras. Med. Trop. 2013, 46, 299–303. [Google Scholar] [CrossRef] [Green Version]
  49. Anderson, G.G.; Kenney, T.F.; Macleod, D.L.; Henig, N.R.; O’Toole, G.A. Eradication of Pseudomonas aeruginosa biofilms on cultured airway cells by a fosfomycin/tobramycin antibiotic combination. Pathog. Dis. 2013, 67, 39–45. [Google Scholar] [CrossRef] [Green Version]
  50. Yu, W.; Zhou, K.; Guo, L.; Ji, J.; Niu, T.; Xiao, T.; Shen, P.; Xiao, Y. In vitro Pharmacokinetics/Pharmacodynamics Evaluation of Fosfomycin Combined with Amikacin or Colistin against KPC2-Producing Klebsiella pneumoniae. Front. Cell. Infect. Microbiol. 2017, 7, 246. [Google Scholar] [CrossRef] [Green Version]
  51. Rodríguez-Avial, I.; Pena, I.; Picazo, J.J.; Rodríguez-Avial, C.; Culebras, E. In vitro activity of the next-generation aminoglycoside plazomicin alone and in combination with colistin, meropenem, fosfomycin or tigecycline against carbapenemase-producing Enterobacteriaceae strains. Int. J. Antimicrob. Agents 2015, 46, 616–621. [Google Scholar] [CrossRef]
  52. Montgomery, A.B.; Rhomberg, P.R.; Abuan, T.; Walters, K.A.; Flamm, R.K. Potentiation effects of amikacin and fosfomycin against selected amikacin-nonsusceptible Gram-negative respiratory tract pathogens. Antimicrob. Agents Chemother. 2014, 58, 3714–3719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Souli, M.; Galani, I.; Boukovalas, S.; Gourgoulis, M.G.; Chryssouli, Z.; Kanellakopoulou, K.; Panagea, T.; Giamarellou, H. In vitro interactions of antimicrobial combinations with fosfomycin against KPC-2-producing Klebsiella pneumoniae and protection of resistance development. Antimicrob. Agents Chemother. 2011, 55, 2395–2397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Wind, C.M.; De Vries, H.J.C.; Van Dam, A.P. Determination of in vitro synergy for dual antimicrobial therapy against resistant Neisseria gonorrhoeae using Etest and agar dilution. Int. J. Antimicrob. Agents 2015, 45, 305–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Hemapanpairoa, J.; Changpradub, D.; Thunyaharn, S.; Santimaleeworagun, W. Vancomycin-resistant enterococcal infection in a Thai university hospital: Clinical characteristics, treatment outcomes, and synergistic effect. Infect. Drug Resist. 2019, 12, 2049–2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Dinos, G.P. The macrolide antibiotic renaissance. Br. J. Pharmacol. 2017, 174, 2967–2983. [Google Scholar] [CrossRef] [PubMed]
  57. Hauser, C.; Hirzberger, L.; Unemo, M.; Furrer, H.; Endimiani, A. In vitro activity of fosfomycin alone and in combination with ceftriaxone or azithromycin against clinical Neisseria gonorrhoeae isolates. Antimicrob. Agents Chemother. 2015, 59, 1605–1611. [Google Scholar] [CrossRef] [Green Version]
  58. Presterl, E.; Hajdu, S.; Lassnigg, A.M.; Hirschl, A.M.; Holinka, J.; Graninger, W. Effects of azithromycin in combination with vancomycin, daptomycin, fosfomycin, tigecycline, and ceftriaxone on Staphylococcus epidermidis biofilms. Antimicrob. Agents Chemother. 2009, 53, 3205–3210. [Google Scholar] [CrossRef] [Green Version]
  59. Kasai, T.; Yuzuru Homma, J. Synergistic effects of a macrolide and a cell wall-affecting antibiotic on pseudomonas aeruginosa in vitro and in vivo 2. Combined effects of a macrolide with a fosfomycin and an aminoglycoside antibiotic. J. Antibiot. (Tokyo) 1982, 35, 858–865. [Google Scholar] [CrossRef] [Green Version]
  60. Dicicco, M.; Neethirajan, S.; Weese, J.S.; Singh, A. In vitro synergism of fosfomycin and clarithromycin antimicrobials against methicillin-resistant Staphylococcus pseudintermedius. BMC Microbiol. 2014, 14, 129. [Google Scholar] [CrossRef] [Green Version]
  61. Rodriguez, A.; Vicente, M.V.; Olay, T. Single- and combination-antibiotic therapy for experimental endocarditis caused by methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 1987, 31, 1444–1445. [Google Scholar] [CrossRef] [Green Version]
  62. Shi, J.; Mao, N.F.; Wang, L.; Zhang, H.B.; Chen, Q.; Liu, H.; Tang, X.; Jin, T.; Zhu, C.T.; Li, F.B.; et al. Efficacy of combined vancomycin and fosfomycin against methicillin-resistant Staphylococcus aureus in biofilms in vivo. PLoS ONE 2014, 9, e113133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Coronado-Álvarez, N.M.; Parra, D.; Parra-Ruiz, J. Clinical efficacy of fosfomycin combinations against a variety of Gram-positive cocci. Enferm. Infecc. Microbiol. Clin. 2019, 37, 4–10. [Google Scholar] [CrossRef] [PubMed]
  64. Shankar, C.; Nabarro, L.E.B.; Anandan, S.; Veeraraghavan, B. Minocycline and Tigecycline: What Is Their Role in the Treatment of Carbapenem-Resistant Gram-Negative Organisms? Microb. Drug Resist. 2017, 23, 437–446. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, Y.; Chen, F.; Sun, E.; Ma, R.; Qu, C.; Ma, L. In vitro antibacterial activity of combinations of fosfomycin, minocycline and polymyxin B on pan-drug-resistant Acinetobacter baumannii. Exp. Ther. Med. 2013, 5, 1737–1739. [Google Scholar] [CrossRef] [Green Version]
  66. Daza, R.; Moreno Lopez, M.; Damaso, D. Interactions of fosfomycin with other antibiotics. Chemotherapy 1977, 23, 86–92. [Google Scholar] [CrossRef]
  67. Descourouez, J.L.; Jorgenson, M.R.; Wergin, J.E.; Rose, W.E. Fosfomycin synergy in vitro with amoxicillin, daptomycin, and linezolid against vancomycin-resistant enterococcus faecium from renal transplant patients with infected urinary stents. Antimicrob. Agents Chemother. 2013, 57, 1518–1520. [Google Scholar] [CrossRef] [Green Version]
  68. Davis, H.; Brown, R.; Ashcraft, D.; Pankey, G. In Vitro Synergy with Fosfomycin Plus Doxycycline Against Linezolid and Vancomycin-resistant Enterococcus faecium. J. Glob. Antimicrob. Resist. 2020, 22, 78–83. [Google Scholar] [CrossRef]
  69. Tang, H.-J.; Chen, C.-C.; Cheng, K.-C.; Toh, H.-S.; Su, B.-A.; Chiang, S.-R.; Ko, W.-C.; Chuang, Y.-C. In vitro efficacy of fosfomycin-containing regimens against methicillin-resistant Staphylococcus aureus in biofilms. J. Antimicrob. Chemother. 2012, 67, 944–950. [Google Scholar] [CrossRef] [Green Version]
  70. Sun, C.; Falagas, M.E.; Wang, R.; Karageorgopoulos, D.E.; Yu, X.; Liu, Y.; Cai, Y.; Liang, B.; Song, X.; Liu, Z. In vitro activity of minocycline combined with fosfomycin against clinical isolates of methicillin-resistant Staphylococcus aureus. J. Antibiot. (Tokyo) 2011, 64, 559–562. [Google Scholar] [CrossRef] [Green Version]
  71. Crémieux, A.-C.; Dinh, A.; Nordmann, P.; Mouton, W.; Tattevin, P.; Ghout, I.; Jayol, A.; Aimer, O.; Gatin, L.; Verdier, M.-C.; et al. Efficacy of colistin alone and in various combinations for the treatment of experimental osteomyelitis due to carbapenemase-producing Klebsiella pneumoniae. J. Antimicrob. Chemother. 2019, 74, 2666–2675. [Google Scholar] [CrossRef] [Green Version]
  72. Ku, N.S.; Lee, S.H.; Lim, Y.S.; Choi, H.; Ahn, J.Y.; Jeong, S.J.; Shin, S.J.; Choi, J.Y.; Choi, Y.H.; Yeom, J.S.; et al. In vivo efficacy of combination of colistin with fosfomycin or minocycline in a mouse model of multidrug-resistant Acinetobacter baumannii pneumonia. Sci. Rep. 2019, 9, 17127. [Google Scholar] [CrossRef] [PubMed]
  73. Corvec, S.; Tafin, U.F.; Betrisey, B.; Borens, O.; Trampuz, A. Activities of fosfomycin, tigecycline, colistin, and gentamicin against extended-spectrum-lactamase-producing escherichia coli in a foreign-body infection model. Antimicrob. Agents Chemother. 2013, 57, 1421–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Evren, E.; Azap, Ö.K.; Çolakoǧlu, Ş.; Arslan, H. In vitro activity of fosfomycin in combination with imipenem, meropenem, colistin and tigecycline against OXA 48-positive Klebsiella pneumoniae strains. Diagn. Microbiol. Infect. Dis. 2013, 76, 335–338. [Google Scholar] [CrossRef] [PubMed]
  75. Bulman, Z.P.; Zhao, M.; Satlin, M.J.; Chen, L.; Kreiswirth, B.N.; Walsh, T.J.; Nation, R.L.; Li, J.; Tsuji, B.T. Polymyxin B and fosfomycin thwart KPC-producing Klebsiella pneumoniae in the hollow-fibre infection model. Int. J. Antimicrob. Agents 2018, 52, 114–118. [Google Scholar] [CrossRef] [PubMed]
  76. Walsh, C.C.; Landersdorfer, C.B.; McIntosh, M.P.; Peleg, A.Y.; Hirsch, E.B.; Kirkpatrick, C.M.; Bergen, P.J. Clinically relevant concentrations of fosfomycin combined with polymyxin B, tobramycin or ciprofloxacin enhance bacterial killing of Pseudomonas aeruginosa, but do not suppress the emergence of fosfomycin resistance. J. Antimicrob. Chemother. 2016, 71, 2218–2229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Heidary, M.; Khosravi, A.D.; Khoshnood, S.; Nasiri, M.J.; Soleimani, S.; Goudarzi, M. Daptomycin. J. Antimicrob. Chemother. 2018, 73, 1–11. [Google Scholar] [CrossRef]
  78. Daptomycin Breakpoints for Enterococci; 2019. Available online: https://clsi.org/media/3356/mr06ed1_sample.pdf (accessed on 4 August 2020).
  79. García-de-la-Mària, C.; Gasch, O.; García-Gonzalez, J.; Soy, D.; Shaw, E.; Ambrosioni, J.; Almela, M.; Pericàs, J.M.; Tellez, A.; Falces, C.; et al. The Combination of Daptomycin and Fosfomycin Has Synergistic, Potent, and Rapid Bactericidal Activity against Methicillin-Resistant Staphylococcus aureus in a Rabbit Model of Experimental Endocarditis. Antimicrob. Agents Chemother. 2018, 62, e02633-17. [Google Scholar] [CrossRef] [Green Version]
  80. Aktas, G.; Derbentli, S. In vitro activity of daptomycin combinations with rifampicin, gentamicin, fosfomycin and fusidic acid against MRSA strains. J. Glob. Antimicrob. Resist. 2017, 10, 223–227. [Google Scholar] [CrossRef]
  81. Lingscheid, T.; Poeppl, W.; Bernitzky, D.; Veletzky, L.; Kussmann, M.; Plasenzotti, R.; Burgmann, H. Daptomycin plus fosfomycin: A synergistic combination in experimental implant-associated MRSA-osteomyelitis in rats. Antimicrob. Agents Chemother. 2014, 59, 1–16. [Google Scholar] [CrossRef] [Green Version]
  82. Rice, L.B.; Eliopoulos, C.T.; Yao, J.D.C.; Eliopoulos, G.M.; Moellering, R.C. In vivo activity of the combination of daptomycin and fosfomycin compared with daptomycin alone against a strain of Enterococcus faecalis with high-level gentamicin resistance in the rat endocarditis model. Diagn. Microbiol. Infect. Dis. 1992, 15, 173–176. [Google Scholar] [CrossRef]
  83. Leite, G.C.; Oliveira, M.S.; Perdigão-Neto, L.V.; Rocha, C.K.D.; Guimarães, T.; Rizek, C.; Levin, A.S.; Costa, S.F. Antimicrobial combinations against pan- resistant acinetobacter baumannii isolates with different resistance mechanisms. PLoS ONE 2016, 11, e0151270. [Google Scholar] [CrossRef] [PubMed]
  84. Ku, Y.H.; Chen, C.C.; Lee, M.F.; Chuang, Y.C.; Tang, H.J.; Yu, W.L. Comparison of synergism between colistin, fosfomycin and tigecycline against extended-spectrum β-lactamase-producing Klebsiella pneumoniae isolates or with carbapenem resistance. J. Microbiol. Immunol. Infect. 2017, 50, 931–939. [Google Scholar] [CrossRef] [PubMed]
  85. Berçot, B.; Poirel, L.; Dortet, L.; Nordmann, P. In vitro evaluation of antibiotic synergy for NDM-1-producing Enterobacteriaceae. J. Antimicrob. Chemother. 2011, 66, 2295–2297. [Google Scholar] [CrossRef] [PubMed]
  86. Samonis, G.; Maraki, S.; Karageorgopoulos, D.E.; Vouloumanou, E.K.; Falagas, M.E. Synergy of fosfomycin with carbapenems, colistin, netilmicin, and tigecycline against multidrug-resistant Klebsiella pneumoniae, Escherichia coli, and Pseudomonas aeruginosa clinical isolates. Eur. J. Clin. Microbiol. Infect. Dis. 2012, 31, 695–701. [Google Scholar] [CrossRef] [PubMed]
  87. Papoutsaki, V.; Galani, I.; Papadimitriou, E.; Karantani, I.; Karaiskos, I.; Giamarellou, H. Evaluation of in vitro methods for testing tigecycline combinations against carbapenemase-producing Klebsiella pneumoniae isolates. J. Glob. Antimicrob. Resist. 2020, 20, 98–104. [Google Scholar] [CrossRef]
  88. Huang, L.; Wang, M.; Sun, L. Synergy Testing by E-Test and Microdilution Checkerboard for Fosfomycin Combined with Tigecycline against KPC-Producing Klebsiella pneumoniae. Clin. Lab. 2019, 65, 2369–2375. [Google Scholar] [CrossRef]
  89. Yu, W.; Shen, P.; Bao, Z.; Zhou, K.; Zheng, B.; Ji, J.; Guo, L.; Huang, C.; Xiao, Y. In vitro antibacterial activity of fosfomycin combined with other antimicrobials against KPC-producing Klebsiella pneumoniae. Int. J. Antimicrob. Agents 2017, 50, 237–241. [Google Scholar] [CrossRef]
  90. Simonetti, O.; Morroni, G.; Ghiselli, R.; Orlando, F.; Brenciani, A.; Xhuvelaj, L.; Provinciali, M.; Offidani, A.; Guerrieri, M.; Giacometti, A.; et al. In vitro and in vivo activity of fosfomycin alone and in combination with rifampin and tigecycline against Grampositive cocci isolated from surgical wound infections. J. Med. Microbiol. 2018, 67, 139–143. [Google Scholar] [CrossRef]
  91. Tang, H.J.; Chen, C.C.; Cheng, K.C.; Wu, K.Y.; Lin, Y.C.; Zhang, C.C.; Weng, T.C.; Yu, W.L.; Chiu, Y.H.; Toh, H.S.; et al. In Vitro efficacies and resistance profiles of rifampin-based combination regimens for biofilm-embedded methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2013, 57, 5717–5720. [Google Scholar] [CrossRef] [Green Version]
  92. Batts, D. Linezolid--a New Option for Treating Gram-Positive Infections. Oncology (willist. Park.) 2000, 14, 23–29. [Google Scholar] [CrossRef]
  93. Hashemian, S.M.R.; Farhadi, T.; Ganjparvar, M. Linezolid: A review of its properties, function, and use in critical care. Drug Des. Devel. Ther. 2018, 12, 1759–1767. [Google Scholar] [CrossRef] [PubMed]
  94. Chai, D.; Liu, X.; Wang, R.; Bai, Y.; Cai, Y. Efficacy of Linezolid and Fosfomycin in Catheter-Related Biofilm Infection Caused by Methicillin-Resistant Staphylococcus aureus. Biomed Res. Int. 2016, 2016, 6413982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Li, L.; Chen, H.; Liu, Y.; Xu, S.; Wu, M.; Liu, Z.; Qi, C.; Zhang, G.; Li, J.; Huang, X. Synergistic effect of linezolid with fosfomycin against Staphylococcus aureus in vitro and in an experimental Galleria mellonella model. J. Microbiol. Immunol. Infect. 2018, S1684-1182(18)30538-3. [Google Scholar] [CrossRef] [PubMed]
  96. Mihailescu, R.; Tafin, U.F.; Corvec, S.; Oliva, A.; Betrisey, B.; Borens, O.; Trampuza, A. High activity of fosfomycin and rifampin against methicillin-resistant staphylococcus aureus biofilm in vitro and in an experimental foreign-body infection model. Antimicrob. Agents Chemother. 2014, 58, 2547–2553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Blacky, A.; Makristathis, A.; Apfalter, P.; Willinger, B.; Rotter, M.L.; Hirschl, A.M. In vitro activity of fosfomycin alone and in combination with amoxicillin, clarithromycin and metronidazole against Helicobacter pylori compared with combined clarithromycin and metronidazole. Eur. J. Clin. Microbiol. Infect. Dis. 2005, 24, 276–279. [Google Scholar] [CrossRef]
  98. Nord, C.; Lahnborg, G. Efficacy of Metronidazole and Fosfomycin Alone and in Combination in the Treatment of Experimentally Induced Intra-Abdominal Infections. Scand. J. Gastroenterol. Suppl. 1984, 90, 15–19. [Google Scholar] [CrossRef] [Green Version]
  99. Santimaleeworagun, W.; Wongpoowarak, P.; Chayakul, P.; Pattharachayakul, S.; Tansakul, P.; Garey, K.W. In vitro activity of colistin or sulbactam in combination with fosfomycin or imipenem against clinical isolates of carbapenem-resistant acinetobacter baumannii producing OXA-23 carbapenemases. Southeast Asian J. Trop. Med. Public Health 2011, 42, 890–900. [Google Scholar]
  100. Duez, J.M.; Adochitei, A.; Péchinot, A.; Siebor, E.; Sixt, N.; Neuwirth, C. In vitro combinations of five intravenous antibiotics with Dalfopristin-Quinupristin against staphylococcus aureus in a 3-dimensional model. J. Chemother. 2008, 20, 684–689. [Google Scholar] [CrossRef]
  101. Yu, X.H.; Song, X.J.; Cai, Y.; Liang, B.B.; Lin, D.F.; Wang, R. In vitro activity of two old antibiotics against clinical isolates of methicillin-resistant Staphylococcus aureus. J. Antibiot. (Tokyo) 2010, 63, 657–659. [Google Scholar] [CrossRef] [Green Version]
  102. Drugeon, H.; Caillon, J.; Juvin, M. In-vitro Antibacterial Activity of Fusidic Acid Alone and in Combination With Other Antibiotics Against Methicillin-Sensitive and -Resistant Staphylococcus Aureus. J. Antimicrob. Chemother. 1994, 34, 899–907. [Google Scholar] [CrossRef]
  103. Schwarz, S.; Kehrenberg, C.; Doublet, B.; Cloeckaert, A. Molecular basis of bacterial resistance to chloramphenicol and florfenicol. FEMS Microbiol. Rev. 2004, 28, 519–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Perea, E.J.; Torres, M.A.; Borobio, M.V. Synergism of fosfomycin-ampicillin and fosfomycin-chloramphenicol against Salmonella and Shigella. Antimicrob. Agents Chemother. 1978, 13, 705–709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Figueroa, J.; Baquero, G.; Otal, C.; Rodríguez, A. Treatment of typhoid fever with fosfomycin alone and associated to chloramphenicol or ampicillin. Chemotherapy 1977, 23, 365–377. [Google Scholar] [CrossRef] [PubMed]
  106. Masters, P.A.; O’Bryan, T.A.; Zurlo, J.; Miller, D.Q.; Joshi, N. Trimethoprim-sulfamethoxazole revisited. Arch. Intern. Med. 2003, 163, 402–410. [Google Scholar] [CrossRef]
  107. She, P.; Zhou, L.; Li, S.; Liu, Y.; Xu, L.; Chen, L.; Luo, Z.; Wu, Y. Synergistic Microbicidal Effect of Auranofin and Antibiotics Against Planktonic and Biofilm-Encased S. aureus and E. faecalis. Front. Microbiol. 2019, 10, 2453. [Google Scholar] [CrossRef]
  108. Domalaon, R.; Brizuela, M.; Eisner, B.; Findlay, B.; Zhanel, G.G.; Schweizer, F. Dilipid ultrashort cationic lipopeptides as adjuvants for chloramphenicol and other conventional antibiotics against Gram-negative bacteria. Amino Acids 2019, 51, 383–393. [Google Scholar] [CrossRef]
  109. Wang, H.; Wang, H.; Yu, X.; Zhou, H.; Li, B.; Chen, G.; Ye, Z.; Wang, Y.; Cui, X.; Zheng, Y.; et al. Impact of antimicrobial stewardship managed by clinical pharmacists on antibiotic use and drug resistance in a Chinese hospital, 2010–2016: A retrospective observational study. BMJ Open 2019, 9, e026072. [Google Scholar] [CrossRef] [Green Version]
  110. Lyu, Y.; Domalaon, R.; Yang, X.; Schweizer, F. Amphiphilic lysine conjugated to tobramycin synergizes legacy antibiotics against wild-type and multidrug-resistant pseudomonas aeruginosa. Pept. Sci. 2019, 111, e23091. [Google Scholar] [CrossRef]
  111. Akbari-Ayezloy, E.; Hosseini-Jazani, N.; Yousefi, S.; Habibi, N. Eradication of methicillin resistant S. Aureus biofilm by the combined use of fosfomycin and β-chloro-L-alanine. Iran. J. Microbiol. 2017, 9, 1–10. [Google Scholar]
  112. Breidenstein, E.B.M.; Courvalin, P.; Meziane-Cherif, D. Antimicrobial activity of plectasin NZ2114 in combination with cell wall targeting antibiotics against VanA-type enterococcus faecalis. Microb. Drug Resist. 2015, 21, 373–379. [Google Scholar] [CrossRef]
  113. Sakagami, Y.; Komemushi, S.; Tsukamoto, G.; Kondo, H.; Yoshikawa, A.; Muraoka, O. Anti-vRE and anti-MRSA activities of new quinolones and their synergism with commercial antibiotics. Part 2. Biocontrol. Sci. 2008, 13, 103–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Marchese, A.; Bozzolasco, M.; Gualco, L.; Debbia, E.A.; Schito, G.C.; Schito, A.M. Effect of fosfomycin alone and in combination with N-acetylcysteine on E. coli biofilms. Int. J. Antimicrob. Agents 2003, 22, 95–100. [Google Scholar] [CrossRef]
  115. Sakagami, Y.; Mimura, M.; Kajimura, K.; Yokoyama, H.; Iinuma, M.; Tanaka, T.; Ohyama, M. Anti-MRSA activity of sophoraflavanone G and synergism with other antibacterial agents. Lett. Appl. Microbiol. 1998, 27, 98–100. [Google Scholar] [CrossRef] [PubMed]
  116. Dulaney, E.L.; Jacobsen, C.A. Synergy between fosfomycin and arenaemycin. J. Antibiot. (Tokyo) 1988, 41, 982–983. [Google Scholar] [CrossRef]
  117. Zhang, F.; Zhai, T.; Haider, S.; Liu, Y.; Huang, Z.J. Synergistic Effect of Chlorogenic Acid and Caffeic Acid with Fosfomycin on Growth Inhibition of a Resistant Listeria monocytogenes Strain. ACS Omega 2020, 5, 7537–7544. [Google Scholar] [CrossRef] [Green Version]
  118. Abo-Shama, U.H.; El-Gendy, H.; Mousa, W.S.; Hamouda, R.A.; Yousuf, W.E.; Hetta, H.F.; Abdeen, E.E. Synergistic and antagonistic effects of metal nanoparticles in combination with antibiotics against some reference strains of pathogenic microorganisms. Infect. Drug Resist. 2020, 13, 351–362. [Google Scholar] [CrossRef] [Green Version]
  119. Seok, H.; Choi, J.Y.; Wi, Y.M.; Park, D.W.; Peck, K.R.; Ko, K.S. Fosfomycin resistance in escherichia coli isolates from south korea and in vitro activity of fosfomycin alone and in combination with other antibiotics. Antibiotics 2020, 9, 112. [Google Scholar] [CrossRef] [Green Version]
  120. Hickman, R.A.; Hughes, D.; Cars, T.; Malmberg, C.; Cars, O. Cell-wall-inhibiting antibiotic combinations with activity against multidrug-resistant Klebsiella pneumoniae and Escherichia coli. Clin. Microbiol. Infect. 2014, 20, O267–O273. [Google Scholar] [CrossRef] [Green Version]
  121. Borowski, J.; Linda, H. Combined action of fosfomycin with β lactam and aminoglycoside antibiotics. Chemotherapy 1977, 23, 82–85. [Google Scholar] [CrossRef]
  122. Takahashi, K.; Kanno, H. Synergistic activities of combinations of β-lactams, fosfomycin, and tobramycin against Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 1984, 26, 789–791. [Google Scholar] [CrossRef] [Green Version]
  123. Ferrara, A.; Dos Santos, C.; Cimbro, M.; Gialdroni Grassi, G. Effect of different combinations of sparfloxacin, oxacillin, and fosfomycin against methicillin-resistant staphylococci. Eur. J. Clin. Microbiol. Infect. Dis. 1997, 16, 535–537. [Google Scholar] [CrossRef] [PubMed]
  124. Komatsuzawa, H.; Suzuki, J.; Sugai, M.; Miyake, Y.; Suginaka, H. Effect of Combination of Oxacillin and Non-Beta-Lactam Antibiotics on Methicillin-Resistant Staphylococcus Aureus. J. Antimicrob. Chemother. 1994, 33, 1155–1163. [Google Scholar] [CrossRef] [PubMed]
  125. Bañón Arias, R.; García López, M.; Pinedo Sánchez, A. Time–kill evaluation of antimicrobial regimens against clinical isolates of penicillin-resistant Streptococcus pneumoniae. J. Chemother. 2001, 13, 535–540. [Google Scholar] [CrossRef] [PubMed]
  126. Kikuchi, K.; Totsuka, K.; Shimizu, K.; Ishii, T.; Yoshida, T.; Orikasa, Y. Effects of Combination of Benzylpenicillin and Fosfomycin on Penicillin-Resistant Streptococcus pneumoniae. Microb. Drug Resist. 1995, 1, 185–189. [Google Scholar] [CrossRef] [PubMed]
  127. Pestel, M.; Martin, E.; Aucouturier, C.; Lemeland, J.F.; Caron, F. In vitro interactions between different β-lactam antibiotics and fosfomycin against bloodstream isolates of enterococci. Antimicrob. Agents Chemother. 1995, 39, 2341–2344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Farina, C.; Russello, G.; Chinello, P.; Pasticci, M.B.; Raglio, A.; Ravasio, V.; Rizzi, M.; Scarparo, C.; Vailati, F.; Suter, F. In vitro activity effects of twelve antibiotics alone and in association against twenty-seven enterococcus faecalis strains isolated from italian patients with infective endocarditis: High in vitro synergistic effect of the association ceftriaxone-fosfomycin. Chemotherapy 2012, 57, 426–433. [Google Scholar] [CrossRef]
  129. Mullane, E.M.; Avery, L.M.; Nicolau, D.P. Comparative Evaluation of the In Vitro Activities of WCK 5222 (Cefepime-Zidebactam) and Combination Antibiotic Therapies against Carbapenem-Resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2020, 64, e01669-19. [Google Scholar] [CrossRef]
  130. Hayami, H.; Goto, T.; Kawahara, M.; Ohi, Y. Activities of β-lactams, fluoroquinolones, amikacin and fosfomycin alone and in combination against Pseudomonas aeruginosa isolated from complicated urinary tract infections. J. Infect. Chemother. 1999, 5, 130–138. [Google Scholar] [CrossRef]
  131. Tessier, F.; Quentin, C. In vitro activity of fosfomycin combined with ceftazidime, imipenem, amikacin, and ciprofloxacin against Pseudomonas aeruginosa. Eur. J. Clin. Microbiol. Infect. Dis. 1997, 16, 159–162. [Google Scholar] [CrossRef]
  132. Martinez-Martinez, L.; Rodriguez, G.; Pascual, A.; Suàrez, A.; Perea, E.J. In-vitro Activity of Antimicrobial Agent Combinations Against Multiresistant Acinetobacter Baumannii. J. Antimicrob. Chemother. 1996, 38, 1107–1108. [Google Scholar] [CrossRef] [Green Version]
  133. Matsuda, K.; Asahi, Y.; Nakagawa, S.; Tanaka, N.; Inoue, M. In-vitro Activity of Imipenem Combined With Beta-Lactam Antibiotics for Methicillin-Resistant Staphylococcus Aureus. J. Antimicrob. Chemother. 1991, 27, 809–815. [Google Scholar] [CrossRef] [PubMed]
  134. Doit, C.P.; Bonacorsi, S.P.; Fremaux, A.J.; Sissia, G.; Cohen, R.; Geslin, P.L.; Bingen, E.H. In vitro killing activities of antibiotics at clinically achievable concentrations in cerebrospinal fluid against penicillin-resistant Streptococcus pneumoniae isolated from children with meningitis. Antimicrob. Agents Chemother. 1994, 38, 2655–2659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Barakett, V.; Lesage, D.; Delisle, F.; Burghoffer, B.; Richard, G.; Vergez, P.; Petit, J. Synergy of Cefotaxime and Fosfomycin Against Penicillin-Resistant Pneumococci. J. Antimicrob. Chemother. 1993, 31, 105–109. [Google Scholar] [CrossRef] [PubMed]
  136. Barbee, L.A.; Soge, O.O.; Holmes, K.K.; Golden, M.R. In vitro synergy testing of novel antimicrobial combination therapies against Neisseria gonorrhoeae. J. Antimicrob. Chemother. 2014, 69, 1572–1578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Sugathan, S.; Mandal, J. An invitro experimental study of the effect of fosfomycin in combination with amikacin, ciprofloxacin or meropenem on biofilm formation by multidrug-resistant urinary isolates of Escherichia coli. J. Med. Microbiol. 2019, 68, 1699–1706. [Google Scholar] [CrossRef] [PubMed]
  138. Loose, M.; Link, I.; Naber, K.G.; Wagenlehner, F.M.E. Carbapenem-Containing Combination Antibiotic Therapy against Carbapenem-Resistant Uropathogenic Enterobacteriaceae. Antimicrob. Agents Chemother. 2019, 64, e01839-19. [Google Scholar] [CrossRef]
  139. Lingscheid, T.; Tobudic, S.; Poeppl, W.; Mitteregger, D.; Burgmann, H. In vitro activity of doripenem plus fosfomycin against drug-resistant clinical blood isolates. Pharmacology 2013, 91, 214–218. [Google Scholar] [CrossRef]
  140. Netikul, T.; Leelaporn, A.; Leelarasmee, A.; Kiratisin, P. In vitro activities of fosfomycin and carbapenem combinations against carbapenem non-susceptible Escherichia coli and Klebsiella pneumoniae. Int. J. Antimicrob. Agents 2010, 35, 609–610. [Google Scholar] [CrossRef]
  141. Bakthavatchalam, Y.D.; Shankar, A.; Muthuirulandi Sethuvel, D.P.; Asokan, K.; Kanthan, K.; Veeraraghavan, B. Synergistic activity of fosfomycin-meropenem and fosfomycin-colistin against carbapenem resistant Klebsiella pneumoniae: An in vitro evidence. Futur. Sci. OA 2020, 6, FSO461. [Google Scholar] [CrossRef]
  142. Erturk Sengel, B.; Altinkanat Gelmez, G.; Soyletir, G.; Korten, V. In vitro synergistic activity of fosfomycin in combination with meropenem, amikacin and colistin against OXA-48 and/or NDM-producing Klebsiella pneumoniae. J. Chemother. 2020, 32, 1–7. [Google Scholar] [CrossRef]
  143. Perdigão Neto, L.V.; Oliveira, M.S.; Martins, R.C.R.; Marchi, A.P.; Gaudereto, J.J.; da Costa, L.A.T.J.; de Lima, L.F.A.; Takeda, C.F.V.; Costa, S.F.; Levin, A.S. Fosfomycin in severe infections due to genetically distinct pan-drug-resistant Gram-negative microorganisms: Synergy with meropenem. J. Antimicrob. Chemother. 2019, 74, 177–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tseng, S.-P.; Wang, S.-F.; Ma, L.; Wang, T.-Y.; Yang, T.-Y.; Siu, L.K.; Chuang, Y.-C.; Lee, P.-S.; Wang, J.-T.; Wu, T.-L.; et al. The plasmid-mediated fosfomycin resistance determinants and synergy of fosfomycin and meropenem in carbapenem-resistant Klebsiella pneumoniae isolates in Taiwan. J. Microbiol. Immunol. Infect. 2017, 50, 653–661. [Google Scholar] [CrossRef] [PubMed]
  145. Albiero, J.; Sy, S.K.B.; Mazucheli, J.; Caparroz-Assef, S.M.; Costa, B.B.; Alves, J.L.B.; Gales, A.C.; Tognim, M.C.B. Pharmacodynamic evaluation of the potential clinical utility of fosfomycin and meropenem in combination therapy against KPC-2-producing Klebsiella pneumoniae. Antimicrob. Agents Chemother. 2016, 60, 4128–4139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Tängdén, T.; Hickman, R.A.; Forsberg, P.; Lagerbäck, P.; Giske, C.G.; Cars, O. Evaluation of double- and triple-antibiotic combinations for VIM- and NDM-producing klebsiella pneumoniae by in vitro time–kill experiments. Antimicrob. Agents Chemother. 2014, 58, 1757–1762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. 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-lactamase. Antimicrob. Agents Chemother. 2019, 63, e00126-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Drusano, G.L.; Neely, M.N.; Yamada, W.M.; Duncanson, B.; Brown, D.; Maynard, M.; Vicchiarelli, M.; Louie, A. The Combination of Fosfomycin plus Meropenem Is Synergistic for Pseudomonas aeruginosa PAO1 in a Hollow-Fiber Infection Model. Antimicrob. Agents Chemother. 2018, 62, e01682-18. [Google Scholar] [CrossRef] [Green Version]
  149. Hamou-Segarra, M.; Zamorano, L.; Vadlamani, G.; Chu, M.; Sanchez-Diener, I.; Juan, C.; Blazquez, J.; Hattie, M.; Stubbs, K.A.; Mark, B.L.; et al. Synergistic activity of fosfomycin, β-lactams and peptidoglycan recycling inhibition against Pseudomonas aeruginosa. J. Antimicrob. Chemother. 2017, 72, 448–454. [Google Scholar] [CrossRef]
  150. Kunakonvichaya, B.; Thirapanmethee, K.; Khuntayaporn, P.; Montakantikul, P.; Chomnawang, M.T. Synergistic effects of fosfomycin and carbapenems against carbapenem-resistant Pseudomonas aeruginosa clinical isolates. Int. J. Antimicrob. Agents 2015, 45, 556–557. [Google Scholar] [CrossRef]
  151. Zhu, W.; Wang, Y.; Cao, W.; Cao, S.; Zhang, J. In vitro evaluation of antimicrobial combinations against imipenem-resistant Acinetobacter baumannii of different MICs. J. Infect. Public Health 2018, 11, 856–860. [Google Scholar] [CrossRef]
  152. Singkham-in, U.; Chatsuwan, T. In vitro activities of carbapenems in combination with amikacin, colistin, or fosfomycin against carbapenem-resistant Acinetobacter baumannii clinical isolates. Diagn. Microbiol. Infect. Dis. 2018, 91, 169–174. [Google Scholar] [CrossRef]
  153. Guggenbichler, J.P.; Berchtold, D.; Allerberger, F.; Bonatti, H.; Hager, J.; Pfaller, W.; Dierich, M.P. In vitro and in vivo effect of antibiotics on catheters colonized by staphylococci. Eur. J. Clin. Microbiol. Infect. Dis. 1992, 11, 408–415. [Google Scholar] [CrossRef] [PubMed]
  154. Debbia, E.; Pesce, A.; Schito, G.C. In vitro interactions between teicoplanin and other antibiotics against enterococci and staphylococci. J. Hosp. Infect. 1986, 7, 73–77. [Google Scholar] [CrossRef]
  155. Wang, L.; Di Luca, M.; Tkhilaishvili, T.; Trampuz, A.; Gonzalez Moreno, M. Synergistic Activity of Fosfomycin, Ciprofloxacin, and Gentamicin Against Escherichia coli and Pseudomonas aeruginosa Biofilms. Front. Microbiol. 2019, 10, 2522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Liu, Y.; Li, H.; Zhang, Y.; Ye, Y.; Gao, Y.; Li, J. In vitro and in vivo activity of ciprofloxacin/ fosfomycin combination therapy against ciprofloxacin-resistant Shigella flexneri isolates. Infect. Drug Resist. 2019, 12, 1619–1628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Yamada, S.; Hyo, Y.; Ohmori, S.; Ohuchi, M. Role of ciprofloxacin in its synergistic effect with fosfomycin on drug-resistant strains of Pseudomonas aeruginosa. Chemotherapy 2007, 53, 202–209. [Google Scholar] [CrossRef]
  158. Monden, K.; Ando, E.; Iida, M.; Kumon, H. Role of fosfomycin in a synergistic combination with ofloxacin against Pseudomonas aeruginosa growing in a biofilm. J. Infect. Chemother. 2002, 8, 218–226. [Google Scholar] [CrossRef]
  159. Kumon, H.; Ono, N.; Iida, M.; Nickel, J.C. Combination effect of fosfomycin and ofloxacin against Pseudomonas aeruginosa growing in a biofilm. Antimicrob. Agents Chemother. 1995, 39, 1038–1044. [Google Scholar] [CrossRef] [Green Version]
  160. Xiong, Y.Q.; Potel, G.; Caillon, J.; Stephant, G.; Jehl, F.; Bugnon, D.; Le Conte, P.; Baron, D.; Drugeon, H. Comparative efficacies of ciprofloxacin and pefloxacin alone or in combination with fosfomycin in experimental endocarditis induced by multidrug-susceptible and -resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 1995, 39, 496–499. [Google Scholar] [CrossRef] [Green Version]
  161. Vogt, K.; Hahn, H. Synergism between Ciprofloxacin and Fosfomycin against Gram-negative Bacteria in vitro. Zentralblatt fur Bakteriol. 1989, 272, 225–230. [Google Scholar] [CrossRef]
  162. Figueredo, V.; Neu, H. Synergy of Ciprofloxacin With Fosfomycin in Vitro Against Pseudomonas Isolates From Patients With Cystic Fibrosis. J. Antimicrob. Chemother. 1988, 22, 41–50. [Google Scholar] [CrossRef]
  163. Weber, P.; Boussougant, Y.; Ichou, F.; Dutoit, C.; Carbon, C. Bactericidal Effect of Ofloxacin Alone and Combined With Fosfomycin or Vancomycin Against Staphylococcus Aureus in Vitro and in Sera From Volunteers. J. Antimicrob. Chemother. 1987, 20, 839–847. [Google Scholar] [CrossRef] [PubMed]
  164. Yu, W.; Luo, Q.; Shi, Q.; Huang, C.; Yu, X.; Niu, T.; Zhou, K.; Zhang, J.; Xiao, Y. In vitro antibacterial effect of fosfomycin combination therapy against colistin-resistant Klebsiella pneumoniae. Infect. Drug Resist. 2018, 11, 577–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Li Bassi, G.; Motos, A.; Fernandez-Barat, L.; Aguilera Xiol, E.; Chiurazzi, C.; Senussi, T.; Saco, M.A.; Fuster, C.; Carbonara, M.; Bobi, J.; et al. Nebulized Amikacin and Fosfomycin for Severe Pseudomonas aeruginosa Pneumonia: An Experimental Study. Crit. Care Med. 2019, 47, e470–e477. [Google Scholar] [CrossRef] [PubMed]
  166. Díez-Aguilar, M.; Morosini, M.I.; Tedim, A.P.; Rodríguez, I.; Aktaş, Z.; Cantón, R. Antimicrobial activity of fosfomycin-tobramycin combination against Pseudomonas aeruginosa isolates assessed by time–kill assays and mutant prevention concentrations. Antimicrob. Agents Chemother. 2015, 59, 6039–6045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. McCaughey, G.; McKevitt, M.; Elborn, J.S.; Tunney, M.M. Antimicrobial activity of fosfomycin and tobramycin in combination against cystic fibrosis pathogens under aerobic and anaerobic conditions. J. Cyst. Fibros. 2012, 11, 163–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Cai, Y.; Fan, Y.; Wang, R.; An, M.-M.; Liang, B.-B. Synergistic effects of aminoglycosides and fosfomycin on Pseudomonas aeruginosa in vitro and biofilm infections in a rat model. J. Antimicrob. Chemother. 2009, 64, 563–566. [Google Scholar] [CrossRef] [Green Version]
  169. Chinwuba, Z.G.; Chiori, C.O.; Ghobashy, A.A.; Okore, V.C. Determination of the synergy of antibiotic combinations by an overlay inoculum susceptibility disc method. Arzneimittelforschung 1991, 41, 148–150. [Google Scholar]
  170. López Díaz, M.C.; Ríos, E.; Rodríguez-Avial, I.; Simaluiza, R.J.; Picazo, J.J.; Culebras, E. In-vitro activity of several antimicrobial agents against methicillin-resistant Staphylococcus aureus (MRSA) isolates expressing aminoglycoside-modifying enzymes: Potency of plazomicin alone and in combination with other agents. Int. J. Antimicrob. Agents 2017, 50, 191–196. [Google Scholar] [CrossRef]
  171. Morikawa, K.; Nonaka, M.; Yoshikawa, Y.; Torii, I. Synergistic effect of fosfomycin and arbekacin on a methicillin-resistant Staphylococcus aureus-induced biofilm in a rat model. Int. J. Antimicrob. Agents 2005, 25, 44–50. [Google Scholar] [CrossRef]
  172. Kono, K.; Takeda, S.; Tatara, I.; Arakawa, K. In vitro activities of arbekacin, alone and in combination, against methicillin-resistant Staphylococcus aureus. Jpn. J. Antibiot. 1994, 47, 710–719. [Google Scholar]
  173. MacLeod, D.L.; Barker, L.M.; Sutherland, J.L.; Moss, S.C.; Gurgel, J.L.; Kenney, T.F.; Burns, J.L.; Baker, W.R. Antibacterial activities of a fosfomycin/tobramycin combination: A novel inhaled antibiotic for bronchiectasis. J. Antimicrob. Chemother. 2009, 64, 829–836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Xu, X.; Xu, L.; Yuan, G.; Wang, Y.; Qu, Y.; Zhou, M. Synergistic combination of two antimicrobial agents closing each other’s mutant selection windows to prevent antimicrobial resistance. Sci. Rep. 2018, 8, 7237. [Google Scholar] [CrossRef] [PubMed]
  175. Tang, H.J.; Chen, C.C.; Ko, W.C.; Yu, W.L.; Chiang, S.R.; Chuang, Y.C. In vitro efficacy of antimicrobial agents against high-inoculum or biofilm-embedded meticillin-resistant Staphylococcus aureus with vancomycin minimal inhibitory concentrations equal to 2 μg/mL (VA2-MRSA). Int. J. Antimicrob. Agents 2011, 38, 46–51. [Google Scholar] [CrossRef] [PubMed]
  176. Pistella, E.; Falcone, M.; Baiocchi, P.; Pompeo, M.E.; Perciaccante, A.; Penni, A.; Venditti, M. In vitro activity of fosfomycin in combination with vancomycin or teicoplanin against Staphylococcus aureus isolated from device-associated infections unresponsive to glycopeptide therapy. Infez. Med. 2005, 13, 97–102. [Google Scholar] [PubMed]
  177. Gatermann, S.; Marre, R.; Schulz, E. The microbiological efficacy of the combination of fosfomycin and vancomycin against clinically relevant staphylococci. Infection 1989, 17, 35–37. [Google Scholar] [CrossRef] [PubMed]
  178. Debbia, E.; Varaldo, P.E.; Schito, G.C. In vitro activity of imipenem against enterococci and staphylococci and evidence for high rates of synergism with teicoplanin, fosfomycin, and rifampin. Antimicrob. Agents Chemother. 1986, 30, 813–815. [Google Scholar] [CrossRef] [Green Version]
  179. Gaillard, J.L.; Merlino, R.; Pajot, N.; Goulet, O.; Fauchere, J.L.; Ricour, C.; Veron, M. Conventional and nonconventional modes of vancomycin administration to decontaminate the internal surface of catheters colonized with coagulase-negative staphylococci. J. Parenter. Enter. Nutr. 1990, 14, 593–597. [Google Scholar] [CrossRef]
  180. Simon, V.; Simon, M. Antibacterial Activity of Teicoplanin and Vancomycin in Combination with Rifampicin, Fusidic Acid or Fosfomycin Against Staphylococci on Vein Catheters. Scand. J. Infect. Dis. 1990, 72, 14–19. [Google Scholar] [CrossRef]
  181. Albur, M.S.; Noel, A.; Bowker, K.; MacGowan, A. The combination of colistin and fosfomycin is synergistic against NDM-1-producing Enterobacteriaceae in in vitro pharmacokinetic/pharmacodynamic model experiments. Int. J. Antimicrob. Agents 2015, 46, 560–567. [Google Scholar] [CrossRef]
  182. Wistrand-Yuen, P.; Olsson, A.; Skarp, K.P.; Friberg, L.E.; Nielsen, E.I.; Lagerbäck, P.; Tängdén, T. Evaluation of polymyxin B in combination with 13 other antibiotics against carbapenemase-producing Klebsiella pneumoniae in time-lapse microscopy and time–kill experiments. Clin. Microbiol. Infect. 2020, S1198-743X(20)30149-X. [Google Scholar] [CrossRef] [Green Version]
  183. Wang, J.; He, J.-T.; Bai, Y.; Wang, R.; Cai, Y. Synergistic Activity of Colistin/Fosfomycin Combination against Carbapenemase-Producing Klebsiella pneumoniae in an In Vitro Pharmacokinetic/Pharmacodynamic Model. Biomed Res. Int. 2018, 2018, 5720417. [Google Scholar] [CrossRef] [PubMed]
  184. Di, X.; Wang, R.; Liu, B.; Zhang, X.; Ni, W.; Wang, J.; Liang, B.; Cai, Y.; Liu, Y. Thymidine-Dependent Staphylococcus aureus Small-Colony Variants Are Induced by Trimethoprim-Sulfamethoxazole (SXT) and Have Increased Fitness during SXT Challenge. Antimicrob. Agents Chemother. 2015, 68, 551–555. [Google Scholar] [CrossRef]
  185. Sertcelik, A.; Baran, I.; Akinci, E.; Mumcuoglu, I.; Bodur, H. Synergistic Activities of Colistin Combinations with Meropenem, Sulbactam, Minocycline, Disodium Fosfomycin, or Vancomycin Against Different Clones of Carbapenem-Resistant Acinetobacter baumannii Strains. Microb. Drug Resist. 2020, 26, 429–433. [Google Scholar] [CrossRef] [PubMed]
  186. Bian, X.; Liu, X.; Chen, Y.; Chen, D.; Li, J.; Zhang, J. Dose Optimization of Colistin Combinations against Carbapenem-Resistant Acinetobacter baumannii from Patients with Hospital-Acquired Pneumonia in China by Using an In Vitro Pharmacokinetic/Pharmacodynamic Model. Antimicrob. Agents Chemother. 2019, 63, e01989-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Leelasupasri, S.; Santimaleeworagun, W.; Jitwasinkul, T. Antimicrobial Susceptibility among Colistin, Sulbactam, and Fosfomycin and a Synergism Study of Colistin in Combination with Sulbactam or Fosfomycin against Clinical Isolates of Carbapenem-Resistant Acinetobacter baumannii. J. Pathog. 2018, 2018, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Lertsrisatit, Y.; Santimaleeworagun, W.; Thunyaharn, S.; Traipattanakul, J. In vitro activity of colistin mono- and combination therapy against colistin-resistant Acinetobacter baumannii, mechanism of resistance, and clinical outcomes of patients infected with colistinresistant A. baumannii at a Thai university hospital. Infect. Drug Resist. 2017, 10, 437–443. [Google Scholar] [CrossRef] [Green Version]
  189. Fan, B.; Guan, J.; Wang, X.; Cong, Y. Activity of colistin in combination with meropenem, tigecycline, fosfomycin, fusidic acid, rifampin or sulbactam against extensively drug-resistant acinetobacter baumannii in a murine thigh-infection model. PLoS ONE 2016, 11, e0157757. [Google Scholar] [CrossRef]
  190. Wei, W.; Yang, H.; Liu, Y.; Ye, Y.; Li, J. In vitro synergy of colistin combinations against extensively drug-resistant Acinetobacter baumannii producing OXA-23 carbapenemase. J. Chemother. 2016, 28, 159–163. [Google Scholar] [CrossRef]
  191. Lee, Y.-C.; Chen, P.-Y.; Wang, J.-T.; Chang, S.-C. A study on combination of daptomycin with selected antimicrobial agents: In vitro synergistic effect of MIC value of 1 mg/L against MRSA strains. BMC Pharmacol. Toxicol. 2019, 20, 25. [Google Scholar] [CrossRef] [Green Version]
  192. Miŕo, J.M.; Entenza, J.M.; Del Río, A.; Velasco, M.; Castañeda, X.; De La Mària, C.G.; Giddey, M.; Armero, Y.; Pericàs, J.M.; Cervera, C.; et al. High-dose daptomycin plus fosfomycin is safe and effective in treating methicillin-susceptible and methicillin-resistant Staphylococcus aureus endocarditis. Antimicrob. Agents Chemother. 2012, 56, 4511–4515. [Google Scholar] [CrossRef] [Green Version]
  193. Poeppl, W.; Tobudic, S.; Lingscheid, T.; Plasenzotti, R.; Kozakowski, N.; Lagler, H.; Georgopoulos, A.; Burgmann, H. Daptomycin, fosfomycin, or both for treatment of methicillin-resistant Staphylococcus aureus osteomyelitis in an experimental rat model. Antimicrob. Agents Chemother. 2011, 55, 4999–5003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Zheng, J.X.; Sun, X.; Lin, Z.W.; Qi, G.B.; Tu, H.P.; Wu, Y.; Jiang, S.B.; Chen, Z.; Deng, Q.W.; Qu, D.; et al. In vitro activities of daptomycin combined with fosfomycin or rifampin on planktonic and adherent linezolid-resistant isolates of Enterococcus faecalis. J. Med. Microbiol. 2019, 68, 493–502. [Google Scholar] [CrossRef] [PubMed]
  195. Rice, L.B.; Eliopoulos, G.M.; Moellering, R.C. In vitro synergism between daptomycin and fosfomycin against Enterococcus faecalis isolates with high-level gentamicin resistance. Antimicrob. Agents Chemother. 1989, 33, 470–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Hasse, B.; Husmann, L.; Zinkernagel, A.; Weber, R.; Lachat, M.; Mayer, D. Vascular graft infections. Swiss Med. Wkly. 2013, 143, w13754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Debbia, E.; Pesce, A.; Schito, G.C. In vitro activity of LY146032 alone and in combination with other antibiotics against Gram-positive bacteria. Antimicrob. Agents Chemother. 1988, 32, 279–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Chen, H.; Li, L.; Liu, Y.; Wu, M.; Xu, S.; Zhang, G.; Qi, C.; Du, Y.; Wang, M.; Li, J.; et al. In vitro activity and post-antibiotic effects of linezolid in combination with fosfomycin against clinical isolates of Staphylococcus aureus. Infect. Drug Resist. 2018, 11, 2107–2115. [Google Scholar] [CrossRef] [Green Version]
  199. Xu-Hong, Y.; Falagas, M.E.; Dong, W.; Karageorgopoulos, D.E.; De-Feng, L.; Rui, W. In vitro activity of fosfomycin in combination with linezolid against clinical isolates of methicillin-resistant Staphylococcus aureus. J. Antibiot. (Tokyo) 2014, 67, 369–371. [Google Scholar] [CrossRef]
  200. Sahuquillo Arce, J.M.; Colombo Gainza, E.; Gil Brusola, A.; Ortiz Estévez, R.; Cantón, E.; Gobernado, M. In vitro activity of linezolid in combination with doxycycline, fosfomycin, levofloxacin, rifampicin and vancomycin against methicillin-susceptible Staphylococcus aureus. Rev. Esp. Quimioter. 2006, 19, 252–257. [Google Scholar]
  201. Qi, C.; Xu, S.; Wu, M.; Zhu, S.; Liu, Y.; Huang, H.; Zhang, G.; Li, J.; Huang, X. Pharmacodynamics Of Linezolid-Plus-Fosfomycin Against Vancomycin-Susceptible And -Resistant Enterococci In Vitro And In Vivo Of A Galleria mellonella Larval Infection Model. Infect. Drug Resist. 2019, 12, 3497–3505. [Google Scholar] [CrossRef] [Green Version]
  202. Traub, W.H.; Spohr, M.; Bauer, D. Gentamicin-and methicillin-resistant, clinical isolates of staphylococcus aureus: Comparative in vitro and in vivo efficacy of alternative antimicrobial drugs. Chemotherapy 1984, 30, 102–112. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Antonello, R.M.; Principe, L.; Maraolo, A.E.; Viaggi, V.; Pol, R.; Fabbiani, M.; Montagnani, F.; Lovecchio, A.; Luzzati, R.; Di Bella, S. Fosfomycin as Partner Drug for Systemic Infection Management. A Systematic Review of Its Synergistic Properties from In Vitro and In Vivo Studies. Antibiotics 2020, 9, 500. https://doi.org/10.3390/antibiotics9080500

AMA Style

Antonello RM, Principe L, Maraolo AE, Viaggi V, Pol R, Fabbiani M, Montagnani F, Lovecchio A, Luzzati R, Di Bella S. Fosfomycin as Partner Drug for Systemic Infection Management. A Systematic Review of Its Synergistic Properties from In Vitro and In Vivo Studies. Antibiotics. 2020; 9(8):500. https://doi.org/10.3390/antibiotics9080500

Chicago/Turabian Style

Antonello, Roberta Maria, Luigi Principe, Alberto Enrico Maraolo, Valentina Viaggi, Riccardo Pol, Massimiliano Fabbiani, Francesca Montagnani, Antonio Lovecchio, Roberto Luzzati, and Stefano Di Bella. 2020. "Fosfomycin as Partner Drug for Systemic Infection Management. A Systematic Review of Its Synergistic Properties from In Vitro and In Vivo Studies" Antibiotics 9, no. 8: 500. https://doi.org/10.3390/antibiotics9080500

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