Next Article in Journal
Prevalence, Genetic Homogeneity, and Antibiotic Resistance of Pathogenic Yersinia enterocolitica Strains Isolated from Slaughtered Pigs in Bulgaria
Next Article in Special Issue
Voriconazole Pharmacokinetics in Critically Ill Patients and Extracorporeal Membrane Oxygenation Support: A Retrospective Comparative Case-Control Study
Previous Article in Journal
Tobacco Seed-Based Oral Vaccination against Verocytotoxic O138 Escherichia coli as Alternative Approach to Antibiotics in Weaned Piglets
Previous Article in Special Issue
Population Pharmacokinetic Study of Benzylpenicillin in Critically Unwell Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparing the Synergistic and Antagonistic Interactions of Ciprofloxacin and Levofloxacin Combined with Rifampin against Drug-Resistant Staphylococcus aureus: A Time–Kill Assay

1
Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
2
Asia Pacific Foundation for Infectious Diseases (APFID), Seoul 06351, Republic of Korea
3
Center for Infection Prevention and Control, Samsung Medical Center, Seoul 06351, Republic of Korea
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(4), 711; https://doi.org/10.3390/antibiotics12040711
Submission received: 3 March 2023 / Revised: 30 March 2023 / Accepted: 31 March 2023 / Published: 6 April 2023

Abstract

:
Background: Treatment of device-related infections by drug-resistant Staphylococcus aureus can be challenging, and combination therapy has been proposed as a potential solution. We compared the effectiveness of levofloxacin–rifampin and ciprofloxacin–rifampin combinations in killing methicillin-resistant S. aureus (MRSA) using a time–kill assay. Methods: We randomly selected 15 vancomycin-susceptible S. aureus (VSSA) strains, 3 vancomycin-intermediate S. aureus (VISA) strains, and 12 heterogeneous VISA (hVISA) strains from the Asian Bacterial Bank. Time–kill experiments were performed in duplicate for each isolate. Viable bacterial counts were determined at 0 h, 4 h, 8 h, and 24 h for the ciprofloxacin– and levofloxacin–rifampin combinations at 1× MIC and 0.5× MIC. We compared synergistic and antagonistic interactions between the two combinations. Results: The viable bacterial count significantly decreased after 24 h of exposure to ciprofloxacin–rifampin and levofloxacin–rifampin combinations, with synergy observed more frequently in isolates exposed to ciprofloxacin–rifampin (43.3%) than levofloxacin–rifampin (20.0%) (p = 0.0082). The synergistic interactions of both combinations were more frequently observed in resistant strains with high MICs of ciprofloxacin (≥16 mg/L) and levofloxacin (≥8 mg/L). Levofloxacin tended to exhibit more frequent antagonistic interactions with rifampin than ciprofloxacin, although there was no statistical difference in antagonism between the two combinations. Conclusions: Our study demonstrated that ciprofloxacin exhibits superior synergistic activity against MRSA strains, including VISA/hVISA, when combined with rifampin compared with levofloxacin. High MICs of fluoroquinolones were found to predict synergism. Our results suggest that ciprofloxacin may be a more effective choice than levofloxacin for combination therapy with rifampin in the treatment of MRSA infections.

1. Introduction

Staphylococcus aureus is a leading pathogen responsible for a range of infections in both community and hospital settings, and its antibiotic resistance is on the rise [1]. Infections related to medical devices, such as prosthetic joints, are becoming increasingly common and are often challenging to treat even with bactericidal antibiotics [2,3]. Therefore, there is a critical need for new therapeutic strategies to combat these infections.
Combination therapy has emerged as a potential solution to tackle device-related infections caused by drug-resistant S. aureus, and many studies have examined the efficacy of combining different antibiotics. In particular, a combination of rifampin plus fluoroquinolone has been recommended for the treatment of staphylococcal prosthetic joint infection [4,5]. However, the benefit of this combination remains unclear. In vitro studies have yielded inconsistent results regarding the synergy of fluoroquinolones and rifampin, and some studies have even reported antagonistic effects [6]. Animal model studies have also produced mixed results regarding the efficacy of these antibiotic combinations in improving treatment outcomes [7,8]. Moreover, there is a lack of clinical evidence to support the combined use of rifampin in the treatment of methicillin-resistant S. aureus (MRSA) infections [9]. Most clinical trials have used beta-lactam or vancomycin in combination with rifampin [10,11,12], and there is currently no conclusive clinical evidence supporting the effectiveness of fluoroquinolone and rifampin combination therapy.
To address this gap in knowledge, our study aimed to investigate the potential synergistic or antagonistic effects of different fluoroquinolones in combination with rifampin and identify more effective treatment strategies for device-related infections caused by drug-resistant S. aureus. We compared the killing effects of the combination of levofloxacin and rifampin versus ciprofloxacin and rifampin against MRSA strains using a time–kill assay.

2. Results

2.1. Antimicrobial Susceptibility

The MICs of rifampin for all MRSA strains ranged from 0.015 to 16 mg/L, with the VISA/hVISA strains exhibiting higher MIC90 values (16 mg/L) compared with VSSA strains (0.015 mg/L) (Table 1). Interestingly, the rate of rifampin resistance was found to be significantly higher in VISA/hVISA strains (40%) compared with VSSA strains (0%) (p = 0.0169).
In terms of ciprofloxacin and levofloxacin susceptibility, the MIC50 and MIC90 values did not show a significant difference between VSSA and VISA/hVISA groups. Similarly, the rates of resistance to ciprofloxacin and levofloxacin were not significantly different between the two groups (66.7% vs. 73.3%). It is worth noting that the MIC50 and MIC90 of levofloxacin were lower than those of ciprofloxacin, even though the resistance rates were equal between ciprofloxacin and levofloxacin (Table 1).

2.2. Time–Kill Kinetics of Single Antibacterial Agents

In the time–kill kinetics experiments, single antibacterial agents alone, except for levofloxacin (1× MIC) at 8 h, did not significantly reduce the viable bacterial count or exhibit bactericidal activity at 4 h, 8 h, and 24 h compared with the initial inoculum (Figure 1 and Figure 2; Supplementary Tables S1 and S2). The bactericidal activity of levofloxacin at 8 h was lost by 24 h, and the viable bacterial count at 24 h returned to the level of the initial inoculum, although the range was wide (Figure 1D and Supplementary Table S1).

2.3. Synergistic Interaction

Compared with single antibacterial agents, the median viable bacterial count (log CFU/mL) significantly decreased at 24 h for ciprofloxacin– and levofloxacin–rifampin combinations, demonstrating synergistic interaction (Figure 1 and Figure 2; Supplementary Tables S1 and S2). The ciprofloxacin–rifampin combination showed synergy at both 1× and 0.5× MIC concentrations, while the levofloxacin–rifampin combination showed synergy at the 1× MIC concentration. There was no statistically significant difference in bacterial count between the two combinations (Figure 3).
In 16 strains, the ciprofloxacin– or levofloxacin–rifampin combinations exhibited synergy as shown in Table 2. Notably, the highest rate of synergy was observed at 1× MIC concentration of rifampin combined with ciprofloxacin with a rate of 43.3%. This rate was significantly higher than that of rifampin combined with levofloxacin, which was only 20.0% (p = 0.0082) (Table 3). Only 1 of the 6 strains that showed a high MIC (16 mg/L) of rifampin exhibited synergy exclusively at 1× MIC concentration of rifampin combined with ciprofloxacin (Table 2). Among the 21 resistant strains with ciprofloxacin MICs of 4 mg/L or higher, 11 (52.4%) showed synergy at 1× MIC concentration of rifampin combined with ciprofloxacin. Interestingly, among the 13 strains that showed synergy with the ciprofloxacin–rifampin combination, only 2 were susceptible to ciprofloxacin (Table 2). We also found no difference in the proportions of strains showing synergy between VSSA and VISA/hVISA. The proportion of strains showing bactericidal activity was highest at 1× MIC concentration of rifampin combined with ciprofloxacin, and the rate was significantly higher than that of the levofloxacin–rifampin combination (36.7% vs. 20.0%, p = 0.0253) (Table 2).

2.4. Antagonistic Interaction

Antagonistic interactions were observed in a total of nine strains when tested with either ciprofloxacin–rifampin or levofloxacin–rifampin combinations (Table 2). Interestingly, antagonism appeared to be more common with the levofloxacin–rifampin combination than with the ciprofloxacin–rifampin combination (16.7% vs. 6.7% at 1× MIC; 16.7% vs. 10.0% at 0.5× MIC); however, this difference was not statistically significant (Table 3). No antagonism was observed in ciprofloxacin-resistant strains, and none of the VISA/hVISA strains showed antagonism at 1× MIC concentration of rifampin combined with ciprofloxacin.

2.5. Predictors of Synergistic and Antagonistic Interactions

Further analysis using multiple logistic regression was conducted to identify predictors of synergistic and antagonistic interactions. The results showed that the combination of rifampin and ciprofloxacin had a synergistic effect 2.7 times more frequently in strains with a ciprofloxacin MIC of 16 mg/l or higher than in other strains (p = 0.0082). In contrast, a rifampin MIC of 16 mg/l or higher was negatively associated with synergism in the ciprofloxacin–rifampin combination (odds ratio, −2.559; p = 0.0478). In the levofloxacin–rifampin combination, a levofloxacin MIC of 8 mg/L or higher was found to predict synergism (odds ratio, 2.785; p = 0.0195). However, no predictors of antagonism for the two antibiotic combinations were found.

3. Discussion

In this study, we analyzed time–kill assay data to confirm the synergistic effect of ciprofloxacin–rifampin and levofloxacin–rifampin on the killing of drug-resistant S. aureus strains. Our findings suggest that ciprofloxacin exhibits a stronger synergistic effect when combined with rifampin than levofloxacin. Specifically, we observed a synergistic effect on the median viable bacterial count in both combinations of ciprofloxacin– and levofloxacin–rifampin at 1× MIC at 24 h. However, a synergistic effect at 0.5× MIC was found only in the ciprofloxacin–rifampin combination. Furthermore, a higher proportion of strains showed synergy with the ciprofloxacin–rifampin combination at 1× MIC than with the levofloxacin–rifampin combination.
Rifampin has been recommended as one of the therapeutic agents for the treatment of staphylococcal infections due to its potent bactericidal activity against S. aureus [4,5,13,14]. It has been shown to be highly effective in treating staphylococcal infections related to foreign bodies [15]. In addition, rifampin has been reported to retain activity against multidrug-resistant MRSA strains, making it a valuable option for the treatment of drug-resistant infections [16]. Notably, its efficacy against staphylococci in biofilm has been demonstrated in vitro, in animal models, and in patients with orthopedic device-related infections [14,17]. However, the use of rifampin as a single agent can lead to the development of resistance, highlighting the importance of combination therapy [13,18].
Fluoroquinolones have been considered the best antibiotics to use in combination with rifampin for the treatment of S. aureus infections [4,5,19,20,21]. However, studies have not shown consistent synergistic effects with the combination of fluoroquinolones and rifampin, and in some studies, antagonism has been reported [6]. Despite this, clinical studies on the combination of rifampin and fluoroquinolones have been conducted with ciprofloxacin, levofloxacin, and moxifloxacin as combination partners [22,23,24,25]. In particular, levofloxacin has often been regarded as an attractive combination partner in many observational studies [23,24], mainly because of its lower MIC against S. aureus strains and lower likelihood of developing resistance compared with ciprofloxacin [26,27].
The present study has addressed a critical knowledge gap concerning the optimal fluoroquinolone–rifampin combination for bacterial killing. The lack of research in this area underscores the importance of our study, which has revealed that the ciprofloxacin–rifampin combination exhibits a superior bacterial killing effect compared with the levofloxacin–rifampin combination. The slight difference in the frequency of antagonistic interaction between levofloxacin and rifampin, though not statistically significant, may provide a possible explanation for the observed difference in effect between the two combinations. This suggests that the use of levofloxacin in combination with rifampin may not be optimal, and that ciprofloxacin may be a better choice in this regard. It is also noteworthy that the rifampin–ciprofloxacin combination was not found to cause antagonism in ciprofloxacin-resistant MRSA strains. This observation highlights the potential benefits of using the rifampin–ciprofloxacin combination in the management of ciprofloxacin-resistant MRSA infections. Moreover, multiple logistic regression analyses identified high MIC values of ciprofloxacin and levofloxacin as predictors of synergistic interaction, which indicates that the effect of these antibiotic combinations may depend on the characteristics of the bacterial strain, such as the MIC values for each antibiotic.
Previous studies have also reported antagonism between rifampin and fluoroquinolone, particularly when combined with levofloxacin, which may lead to clinical hesitation in prescribing combination therapy [8,15,28]. Some animal studies have also shown antagonistic effects when either ciprofloxacin or levofloxacin is combined with rifampin [8,15,29]. The mechanism behind this antagonistic interaction between rifampin and fluoroquinolone is quite complex. It has been suggested that the inhibition of RNA synthesis by rifampin is responsible for abolishing the bactericidal killing activities of fluoroquinolone. This negative interaction is related to the activity against DNA supercoiling, which is a crucial step in the replication of bacterial DNA [15]. Consequently, the combination of rifampin and fluoroquinolone may reduce the effectiveness of fluoroquinolone in killing bacteria by compromising its bactericidal activity through decreased DNA supercoiling.
Although there are concerns about the potential antagonistic effects of combining rifampin and fluoroquinolone, previous studies have shown that this combination therapy can be beneficial in treating device-related infections. A study demonstrated that rifampin can antagonize the bactericidal effects of ciprofloxacin in staphylococci during exponential growth [28]. However, the study also showed that in non-growing cells, ciprofloxacin exhibited an additive bactericidal effect instead of antagonism [28]. This indicates that the effectiveness of this combination therapy may depend on the growth stage of the targeted bacteria. Therefore, caution should be exercised when administering rifampin and fluoroquinolone for therapeutic purposes in non-biofilm-related clinical situations, as the effects of this combination may vary depending on the specific circumstances of the infection.
This study has some limitations that need to be considered. Firstly, we only investigated the interaction of antibiotics at concentrations of 1× MIC and 0.5× MIC, which may not accurately predict interactions at higher concentrations. Secondly, our study only provides in vitro experimental results, and further clinical trials are necessary to draw conclusive recommendations for antibiotic combination therapy.
Despite these limitations, our study is significant in its investigation of the interaction between rifampin and fluoroquinolone in combination for many drug-resistant S. aureus strains, including VISA/hVISA. These strains pose a significant challenge for treatment due to their drug resistance. Moreover, our study’s comparison of the interactions between ciprofloxacin and levofloxacin in combination with rifampin using various statistical analysis methods contributes to our understanding of the potential benefits and limitations of antibiotic combinations. In the co-administration of rifampin and fluoroquinolones, it should be noted that fluoroquinolones can cause aortic aneurysm in certain patients, as warned by the U.S. Food and Drug Administration (FDA) [30], and can cause persistent side effects in muscles and ligaments as warned by the European Medicines Agency (EMA) [31]. In addition, rifampin can reduce the concentration of other drugs through interaction when used in combination with certain drugs [32]. Therefore, this combination therapy, especially for critically ill patients, should be administered with caution, and good pharmacovigilance should be maintained.
In conclusion, our study provides evidence that ciprofloxacin exhibits superior synergistic activity against MRSA strains, including VISA/hVISA, when combined with rifampin compared with levofloxacin. Our study also highlights that the synergistic interactions between ciprofloxacin and rifampin, as well as levofloxacin and rifampin, are more frequently observed in resistant strains with high MICs of both fluoroquinolones. Interestingly, levofloxacin tended to exhibit more frequent antagonistic interactions with rifampin than ciprofloxacin. Overall, our results suggest that ciprofloxacin may be a more effective choice than levofloxacin for combination therapy with rifampin in the treatment of MRSA infections. However, further studies are needed to confirm these findings and determine the optimal strategy for combination therapy. These findings also underscore the importance of considering the potential synergistic or antagonistic interactions between antibiotics when selecting treatment regimens for MRSA infections.

4. Materials and Methods

4.1. Bacterial Isolates, Susceptibility Testing, and Genotyping

We tested a total of 30 MRSA strains. To ensure a comprehensive analysis, we specifically aimed to include strains with vancomycin non-susceptibility. To achieve this, we randomly selected 15 vancomycin-susceptible S. aureus (VSSA) strains, along with 3 vancomycin-intermediate S. aureus (VISA) strains and 12 heterogeneous VISA (hVISA) strains, from the MRSA bacterial collections stocked at the Asian Bacterial Bank (Asia Pacific Foundation for Infectious Diseases, Seoul, Republic of Korea). These collections included strains obtained from a previous nationwide bacteremia study in the Republic of Korea [33].
Species identification and initial susceptibility testing were performed using the VITEK®2 system (bioMérieux, Marcy-l’Étoile, France). We confirmed the initial susceptibility profile by broth microdilution according to Clinical and Laboratory Standards Institute (CLSI) guidelines [34]. S. aureus ATCC 29213 and Enterococcus faecium ATCC 29212 were utilized as control strains. We determined VISA and hVISA by the modified population analysis profile (PAP) method. The area under the curve (AUC) of the PAP graph was calculated and compared with that of reference strain Mu3 (ATCC 700698) [35]. We identified strains as VSSA, hVISA, or VISA based on the AUCtest/AUCMu3 ratio using these criteria: VSSA, <0.9; hVISA, 0.9 to 1.3; and VISA, >1.3. Mu50 (ATCC 700699) and Mu3 (ATCC 700698) were included as reference strains for VISA and hVISA, respectively.
Multilocus sequence typing (MLST) was conducted using polymerase chain reaction (PCR) amplification and sequencing of seven housekeeping genes (arcC, aroE, glpF, gmk, pta, ypi, and yqiL), as previously described [36]. The allelic profiles and sequence types (STs) were assigned according to the MLST web site (http://saureus.mlst.net/ (accessed on 3 April 2023)). Multiplex PCR was conducted for assignment of SCCmec types [37]. We performed spa typing as previously described [38] and using the Ridom SpaServer (http://spaserver.ridom.de (accessed on 3 April 2023)).

4.2. Time–Kill Assay

To determine the killing effects of different antibiotic combinations against drug-resistant S. aureus strains, we performed time–kill assays. Each isolate was tested in duplicate using an inoculum of approximately 5 × 105 CFU/mL in a final volume of 10 mL, as described previously [39]. Each isolate was tested against each antimicrobial agent alone and in combination at concentrations equal to 0.5× and 1× minimum inhibitory concentration (MIC). We specifically focused on combinations of rifampin with either ciprofloxacin or levofloxacin and evaluated their potential for synergy or antagonism. Samples were taken at 0 h, 4 h, 8 h, and 24 h after incubation with the indicated antimicrobials. To determine the number of viable bacterial colonies, dilutions were plated using an automatic spiral plater (Interscience, St. Nom, France), and the plates were incubated for 18 h to 24 h at 37 °C. We used a Scan 500 (Interscience, St. Nom, France) to count the bacterial colonies. An antibiotic-free growth control was included in each experiment. Time–kill curves were constructed by plotting mean colony count (log CFU/mL) versus time. Synergy was defined as a reduction of ≥2 log CFU/mL with the combination compared with the most active single agent and a reduction of ≥2 log CFU/mL below the initial inoculum at 24 h, as previously described [40]. Antagonism was defined as an increase of ≥2 log CFU/mL with the combination compared with the most active single agent at 24 h [40]. Bactericidal activity was defined as a reduction of ≥3 log CFU/mL compared with the initial inoculums [40].

4.3. Statistical Analysis

All statistical analyses were conducted using Stata version 11.2 (StataCorp, College Station, TX, USA) and R software (version 4.1.3). A p value less than 0.05 was considered statistically significant. To compare the differences in the antimicrobial resistance rates between VSSA and VISA/hVISA strains, we used both the Chi-square test and Fisher’s exact test, with Fisher’s exact test being utilized for any cell counts less than 5. To compare the proportion of bacterial strains showing synergy and antagonism between two antibiotic combinations, we used McNemar’s test, which is a statistical test used to compare paired proportions. The viable bacterial counts did not follow a normal distribution, as assessed by Shapiro–Wilk test. Therefore, non-parametric statistical tests were used to compare the medians of viable bacterial count between groups. We performed the Friedman test, followed by the Wilcoxon signed-rank test with Bonferroni adjustment for multiple groups. Multiple logistic regression analysis was performed to identify predictors for synergistic actions of ciprofloxacin– and levofloxacin–rifampin combinations. This analysis included variables such as year of isolation, vancomycin susceptibility, sequence type, SCCmec type, spa type, and levels of resistance to rifampin, ciprofloxacin, and levofloxacin.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antibiotics12040711/s1, Table S1: The median and interquartile range of viable bacterial counts after exposure to antibacterial agents (1× MIC) for a specific time period; Table S2: The median and interquartile range of viable bacterial counts after exposure to antibacterial agents (0.5× MIC) for a specific time period; Figure S1: Antibacterial activity of rifampin combined with ciprofloxacin or levofloxacin against drug-resistant Staphylococcus aureus strains (mean CFU/mL with standard deviation).

Author Contributions

Conceptualization, D.R.C. and Y.R.K.; Methodology, D.R.C., Y.R.K., J.-H.K. and C.-I.K.; Validation, D.R.C., K.H. and S.Y.C.; Formal Analysis, Y.R.K. and D.R.C.; Investigation, Y.R.K. and C.-I.K.; Resources, C.-I.K. and K.R.P.; Data Curation, Y.R.K. and D.R.C.; Writing—Original Draft Preparation, Y.R.K.; Writing—Review & Editing, D.R.C.; Visualization, D.R.C.; Supervision, D.R.C.; Project Adiministration, D.R.C.; Funding Acquisition, D.R.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. 2020R1F1A1067794).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

We have no conflicts of interest to declare.

References

  1. Howden, B.P.; Davies, J.K.; Johnson, P.D.; Stinear, T.P.; Grayson, M.L. Reduced vancomycin susceptibility in Staphylococcus aureus, including vancomycin-intermediate and heterogeneous vancomycin-intermediate strains: Resistance mechanisms, laboratory detection, and clinical implications. Clin. Microbiol. Rev. 2010, 23, 99–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tuon, F.F.; Suss, P.H.; Telles, J.P.; Dantas, L.R.; Borges, N.H.; Ribeiro, V.S.T. Antimicrobial treatment of Staphylococcus aureus biofilms. Antibiotics 2023, 12, 87. [Google Scholar] [CrossRef] [PubMed]
  3. Jacqueline, C.; Caillon, J. Impact of bacterial biofilm on the treatment of prosthetic joint infections. J. Antimicrob. Chemother. 2014, 69, i37–i40. [Google Scholar] [CrossRef] [Green Version]
  4. Osmon, D.R.; Berbari, E.F.; Berendt, A.R.; Lew, D.; Zimmerli, W.; Steckelberg, J.M.; Rao, N.; Hanssen, A.; Wilson, W. Diagnosis and management of prosthetic joint infection: Clinical practice guidelines by the Infectious Diseases Society of America. Clin. Infect. Dis. 2013, 56, e1–e25. [Google Scholar] [CrossRef] [Green Version]
  5. Cortes-Penfield, N.W.; Hewlett, A.L.; Kalil, A.C. Adjunctive rifampin following debridement and implant retention for staphylococcal prosthetic joint infection: Is it effective if not combined with a fluoroquinolone? Open Forum. Infect. Dis. 2022, 9, ofac582. [Google Scholar] [CrossRef] [PubMed]
  6. Forrest, G.N.; Tamura, K. Rifampin combination therapy for nonmycobacterial infections. Clin. Microbiol. Rev. 2010, 23, 14–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Dworkin, R.; Modin, G.; Kunz, S.; Rich, R.; Zak, O.; Sande, M. Comparative efficacies of ciprofloxacin, pefloxacin, and vancomycin in combination with rifampin in a rat model of methicillin-resistant Staphylococcus aureus chronic osteomyelitis. Antimicrob. Agents Chemother. 1990, 34, 1014–1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Kaatz, G.W.; Seo, S.M.; Barriere, S.L.; Albrecht, L.M.; Rybak, M.J. Ciprofloxacin and rifampin, alone and in combination, for therapy of experimental Staphylococcus aureus endocarditis. Antimicrob. Agents Chemother. 1989, 33, 1184–1187. [Google Scholar] [CrossRef] [Green Version]
  9. Brown, N.M.; Goodman, A.L.; Horner, C.; Jenkins, A.; Brown, E.M. Treatment of methicillin-resistant Staphylococcus aureus (MRSA): Updated guidelines from the UK. JAC Antimicrob. Resist. 2021, 3, dlaa114. [Google Scholar] [CrossRef]
  10. Karlsen, E.; Borgen, P.; Bragnes, B.; Figved, W.; Grøgaard, B.; Rydinge, J.; Sandberg, L.; Snorrason, F.; Wangen, H.; Witsøe, E.; et al. Rifampin combination therapy in staphylococcal prosthetic joint infections: A randomized controlled trial. J. Orthop. Surg. Res. 2020, 15, 365. [Google Scholar] [CrossRef]
  11. Ma, H.; Cheng, J.; Peng, L.; Gao, Y.; Zhang, G.; Luo, Z. Adjunctive rifampin for the treatment of Staphylococcus aureus bacteremia with deep infections: A meta-analysis. PLoS ONE 2020, 15, e0230383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Thwaites, G.E.; Scarborough, M.; Szubert, A.; Nsutebu, E.; Tilley, R.; Greig, J.; Wyllie, S.A.; Wilson, P.; Auckland, C.; Cairns, J.; et al. Adjunctive rifampicin for Staphylococcus aureus bacteraemia (ARREST): A multicentre, randomised, double-blind, placebo-controlled trial. Lancet 2018, 391, 668–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Liu, C.; Bayer, A.; Cosgrove, S.E.; Daum, R.S.; Fridkin, S.K.; Gorwitz, R.J.; Kaplan, S.L.; Karchmer, A.W.; Levine, D.P.; Murray, B.E.; et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clin. Infect. Dis. 2011, 52, e18–e55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zimmerli, W.; Sendi, P. Role of rifampin against staphylococcal biofilm infections in vitro, in animal models, and in orthopedic-device-related infections. Antimicrob. Agents Chemother. 2019, 63, e01746-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Murillo, O.; Pachon, M.E.; Euba, G.; Verdaguer, R.; Tubau, F.; Cabellos, C.; Cabo, J.; Gudiol, F.; Ariza, J. Antagonistic effect of rifampin on the efficacy of high-dose levofloxacin in staphylococcal experimental foreign-body infection. Antimicrob. Agents Chemother. 2008, 52, 3681–3686. [Google Scholar] [CrossRef] [Green Version]
  16. Guérillot, R.; da Silva, A.G.; Monk, I.; Giulieri, S.; Tomita, T.; Alison, E.; Porter, J.; Pidot, S.; Gao, W.; Peleg, A.Y.; et al. Convergent evolution driven by rifampin exacerbates the global burden of drug-resistant Staphylococcus aureus. mSphere 2018, 3, e00550-17. [Google Scholar] [CrossRef] [Green Version]
  17. Sanchez Jr, C.J.; Shiels, S.M.; Tennet, D.J.; Hardy, S.K.; Murray, C.K.; Wenke, J.C. Rifamycin derivatives are effective against staphylococcal biofilms in vitro and elutable from PMMA. Clin. Orthop. Relat. Res. 2015, 473, 2874–2884. [Google Scholar] [CrossRef]
  18. Coiffier, G.; Albert, J.D.; Arvieux, C.; Guggenbuhl, P. Optimizing combination rifampin therapy for staphylococcal osteoarticular infections. Jt. Bone Spine 2013, 80, 11–17. [Google Scholar] [CrossRef]
  19. Berdal, J.E.; Skramm, I.; Mowinckel, P.; Gulbrandsen, P.; Bjornholt, J.V. Use of rifampicin and ciprofloxacin combination therapy after surgical debridement in the treatment of early manifestation prosthetic joint infections. Clin. Microbiol. Infect. 2005, 11, 843–845. [Google Scholar] [CrossRef] [Green Version]
  20. Dworkin, R.J.; Lee, B.L.; Sande, M.A.; Chambers, H.F. Treatment of right-sided Staphylococcus aureus endocarditis in intravenous drug users with ciprofloxacin and rifampicin. Lancet 1989, 2, 1071–1073. [Google Scholar] [CrossRef]
  21. Beldman, M.; Löwik, C.; Soriano, A.; Albiach, L.; Zijlstra, W.P.; Knobben, B.A.S.; Jutte, P.; Sousa, R.; Carvalho, A.; Goswami, K.; et al. If, when, and how to use rifampin in acute staphylococcal periprosthetic joint infections, a multicentre observational study. Clin. Infect. Dis. 2021, 73, 1634–1641. [Google Scholar] [CrossRef] [PubMed]
  22. Zimmerli, W.; Widmer, A.F.; Blatter, M.; Frei, R.; Ochsner, P.E. Role of rifampin for treatment of orthopedic implant-related staphylococcal infections: A randomized controlled trial. Foreign-Body Infection (FBI) Study Group. JAMA 1998, 279, 1537–1541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Nguyen, S.; Robineau, O.; Titecat, M.; Blondiaux, N.; Valette, M.; Loiez, C.; Migaud, H.; Senneville, E. Influence of daily dosage and frequency of administration of rifampicin-levofloxacin therapy on tolerance and effectiveness in 154 patients treated for prosthetic joint infections. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 1675–1682. [Google Scholar] [CrossRef]
  24. Lora-Tamayo, J.; Euba, G.; Cobo, J.; Horcajada, J.P.; Soriano, A.; Sandoval, E.; Pigrau, C.; Benito, N.; Falgueras, L.; Granados, A.; et al. Short- versus long- duration levofloxacin plus rifampicin for acute staphylococcal prosthetic joint infection managed with implant retention: A randomised clinical trial. Int. J. Antimicrob. Agents 2016, 48, 310–316. [Google Scholar] [CrossRef]
  25. Wouthuyzen-Bakker, M.; Tornero, E.; Morata, L.; Panday, P.V.N.; Jutte, P.; Bori, G.; Kampinga, G.A.; Soriano, A. Moxifloxacin plus rifampin as an alternative for levofloxacin plus rifampin in the treatment of a prosthetic joint infection with Staphylococcus aureus. Int. J. Antimicrob. Agents 2018, 51, 38–42. [Google Scholar] [CrossRef]
  26. Jones, M.E.; Visser, M.R.; Klootwijk, M.; Heisig, P.; Verhoef, J.; Schmitz, F.-J. Comparative activities of clinafloxacin, grepafloxacin, levofloxacin, moxifloxacin, ofloxacin, sparfloxacin, and trovafloxacin and nonquinolones linozelid, quinupristin-dalfopristin, gentamicin, and vancomycin against clinical isolates of ciprofloxacin-resistant and -susceptible Staphylococcus aureus strains. Antimicrob. Agents Chemother. 1999, 43, 421–423. [Google Scholar]
  27. Entenza, J.M.; Vouillamoz, J.; Glauser, M.P.; Moreillon, P. Levofloxacin versus ciprofloxacin, flucloxacillin, or vancomycin for treatment of experimental endocarditis due to methicillin-susceptible or -resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 1997, 41, 1662–1667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Bahl, D.; Miller, D.A.; Leviton, I.; Gialanella, P.; Wolin, M.J.; Liu, W.; Perkins, R.; Miller, M.H. In vitro activities of ciprofloxacin and rifampin alone and in combination against growing and nongrowing strains of methicillin-susceptible and methicillin-resistant Staphylococcus aureus. Antimirob. Agents Chemother. 1997, 41, 1293–1297. [Google Scholar] [CrossRef] [Green Version]
  29. Chambers, H.F.; Liu, Q.X.; Chow, L.L.; Hackbarth, C. Efficacy of levofloxacin for experimental aortic-valve endocarditis in rabbits infected with viridans group streptococcus or Staphylococcus aureus. Antimicrob. Agents Chemother. 1999, 43, 2742–4276. [Google Scholar] [CrossRef] [Green Version]
  30. FDA in Brief: FDA Warns that Fluoroquinolone Antibiotics Can Cause Aortic Aneurysm in Certain Patients. Available online: https://www.fda.gov/news-events/fda-brief/fda-brief-fda-warns-fluoroquinolone-antibiotics-can-cause-aortic-aneurysm-certain-patients (accessed on 30 March 2023).
  31. Quinolone- and Fluoroquinolone-Containing Medicinal Products. Available online: https://www.ema.europa.eu/en/medicines/human/referrals/quinolone-fluoroquinolone-containing-medicinal-products (accessed on 30 March 2023).
  32. Niemi, M.; Backman, J.T.; Fromm, M.F.; Neuvonen, P.J.; Kivistö, K.T. Pharmacokinetic interactions with rifampicin: Clinical relevance. Clin. Pharmacokinet. 2003, 42, 819–850. [Google Scholar]
  33. Son, J.S.; Song, J.-H.; Ko, K.S.; Yeom, J.S.; Ki, H.K.; Kim, S.-W.; Chang, H.-H.; Ryu, S.Y.; Kim, Y.-S.; Jung, S.-I.; et al. Bloodstream infections and clinical significance of healthcare-associated bacteremia: A multicenter surveillance study in Korean hospitals. J. Korean Med. Sci. 2010, 25, 992–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Clinical and Laboratory Standards Institute (CLSI). Performance Standards for Antimicrobial Susceptibility Testing: Twenty-Ninth Informational Supplement M100-S29; CLSI: Wayne, PA, USA, 2019. [Google Scholar]
  35. Wootton, M.; Howe, R.A.; Hillman, R.; Walsh, T.R.; Bennett, P.M.; MacGowan, A.P. A modified population analysis profile (PAP) method to detect hetero-resistance to vancomycin in Staphylococcus aureus in a UK hospital. J. Antimicrob. Chemother. 2001, 47, 399–403. [Google Scholar] [CrossRef] [PubMed]
  36. Enright, M.C.; Day, N.P.; Davies, C.E.; Peacock, S.J.; Spratt, B.G. Multilocus sequence typing for characterization of methicillin-resistant and methicillin-susceptible clones of Staphylococcus aureus. J. Clin. Microbiol. 2000, 38, 1008–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Oliveira, D.C.; de Lencastre, H. Multiplex PCR strategy for rapid identification of structural types and variants of the mec element in methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2002, 46, 2155–2161. [Google Scholar] [CrossRef] [Green Version]
  38. Harmsen, D.; Claus, H.; White, W.; Rothgänger, J.; Claus, H.; Turnwald, D.; Vogel, U. Typing of methicillin-resistant Staphylococcus aureus in a university hospital setting by using novel software for spa repeat determination and database management. J. Clin. Microbiol. 2003, 41, 5442–5448. [Google Scholar] [CrossRef] [Green Version]
  39. Garcia, L.S. Clinical Microbiology Procedures Handbook, 3rd ed.; American Society of Microbiology: Washington, DC, USA, 2010. [Google Scholar]
  40. Singh, S.R.; Bacon III, A.E.; Young, D.C.; Cough, K.A. In vitro 24-h time-kill studies of vancomycin and linezolid in combination versus methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2009, 53, 4495–4497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Changes in viable bacterial counts over 24 h after exposure to antibacterial agents at a concentration of 1× MIC (minimum inhibitory concentration). (A) Changes in viable bacterial counts after exposure to control. (B) Changes in viable bacterial counts after exposure to rifampin. (C) Changes in viable bacterial counts after exposure to ciprofloxacin. (D) Changes in viable bacterial counts after exposure to levofloxacin. (E) Changes in viable bacterial counts after exposure to rifampin and ciprofloxacin. (F) Changes in viable bacterial counts after exposure to rifampin and levofloxacin.
Figure 1. Changes in viable bacterial counts over 24 h after exposure to antibacterial agents at a concentration of 1× MIC (minimum inhibitory concentration). (A) Changes in viable bacterial counts after exposure to control. (B) Changes in viable bacterial counts after exposure to rifampin. (C) Changes in viable bacterial counts after exposure to ciprofloxacin. (D) Changes in viable bacterial counts after exposure to levofloxacin. (E) Changes in viable bacterial counts after exposure to rifampin and ciprofloxacin. (F) Changes in viable bacterial counts after exposure to rifampin and levofloxacin.
Antibiotics 12 00711 g001
Figure 2. Changes in viable bacterial counts over 24 h after exposure to antibacterial agents at a concentration of 0.5× MIC (minimum inhibitory concentration). (A) Changes in viable bacterial counts after exposure to control. (B) Changes in viable bacterial counts after exposure to rifampin. (C) Changes in viable bacterial counts after exposure to ciprofloxacin. (D) Changes in viable bacterial counts after exposure to levofloxacin. (E) Changes in viable bacterial counts after exposure to rifampin and ciprofloxacin. (F) Changes in viable bacterial counts after exposure to rifampin and levofloxacin.
Figure 2. Changes in viable bacterial counts over 24 h after exposure to antibacterial agents at a concentration of 0.5× MIC (minimum inhibitory concentration). (A) Changes in viable bacterial counts after exposure to control. (B) Changes in viable bacterial counts after exposure to rifampin. (C) Changes in viable bacterial counts after exposure to ciprofloxacin. (D) Changes in viable bacterial counts after exposure to levofloxacin. (E) Changes in viable bacterial counts after exposure to rifampin and ciprofloxacin. (F) Changes in viable bacterial counts after exposure to rifampin and levofloxacin.
Antibiotics 12 00711 g002
Figure 3. Comparison of viable bacterial counts after exposure to different antibacterial agents for a specific time period. * Adjusted p < 0.0001. (A) Changes in viable bacterial counts following a 4-h exposure to antibacterial agents at a concentration equal to 1× minimum inhibitory concentration (MIC). (B) Changes in viable bacterial counts following a 4-h exposure to antibacterial agents at a concentration equal to 0.5× minimum inhibitory concentration (MIC). (C) Changes in viable bacterial counts following a 8-h exposure to antibacterial agents at a concentration equal to 1× minimum inhibitory concentration (MIC). (D) Changes in viable bacterial counts following a 8-h exposure to antibacterial agents at a concentration equal to 0.5× minimum inhibitory concentration (MIC). (E) Changes in viable bacterial counts following a 24-h exposure to antibacterial agents at a concentration equal to 1× minimum inhibitory concentration (MIC). (F) Changes in viable bacterial counts following a 24-h exposure to antibacterial agents at a concentration equal to 0.5× minimum inhibitory concentration (MIC).
Figure 3. Comparison of viable bacterial counts after exposure to different antibacterial agents for a specific time period. * Adjusted p < 0.0001. (A) Changes in viable bacterial counts following a 4-h exposure to antibacterial agents at a concentration equal to 1× minimum inhibitory concentration (MIC). (B) Changes in viable bacterial counts following a 4-h exposure to antibacterial agents at a concentration equal to 0.5× minimum inhibitory concentration (MIC). (C) Changes in viable bacterial counts following a 8-h exposure to antibacterial agents at a concentration equal to 1× minimum inhibitory concentration (MIC). (D) Changes in viable bacterial counts following a 8-h exposure to antibacterial agents at a concentration equal to 0.5× minimum inhibitory concentration (MIC). (E) Changes in viable bacterial counts following a 24-h exposure to antibacterial agents at a concentration equal to 1× minimum inhibitory concentration (MIC). (F) Changes in viable bacterial counts following a 24-h exposure to antibacterial agents at a concentration equal to 0.5× minimum inhibitory concentration (MIC).
Antibiotics 12 00711 g003
Table 1. MIC distribution of antimicrobials against methicillin-resistant Staphylococcus aureus strains.
Table 1. MIC distribution of antimicrobials against methicillin-resistant Staphylococcus aureus strains.
RifampinCiprofloxacinLevofloxacin
MIC50
(mg/L)
MIC90
(mg/L)
Resistance Rate (%)MIC50
(mg/L)
MIC90
(mg/L)
Resistance Rate (%)MIC50
(mg/L)
MIC90
(mg/L)
Resistance Rate (%)
VSSA
(n = 15)
0.0150.015032>6466.783266.7
VISA/hVISA
(n = 15)
0.0151640.016>6473.383273.3
MIC, minimum inhibitory concentration; VSSA, vancomycin-susceptible S. aureus; VISA, vancomycin-intermediate S. aureus; hVISA, heterogeneous VISA.
Table 2. Microbiological and molecular characteristics of methicillin-resistant Staphylococcus aureus strains and comparison of rifampin interactions with ciprofloxacin and levofloxacin.
Table 2. Microbiological and molecular characteristics of methicillin-resistant Staphylococcus aureus strains and comparison of rifampin interactions with ciprofloxacin and levofloxacin.
IsolatePhenotypeYearSTSCCmec
Type
spa
Type
MIC (mg/L)InteractionBactericidal Activity
0.5× MIC1× MIC0.5× MIC1× MIC
RCLR + CR + LR + CR + LR + CR + LR + CR + L
1VSSA2006239IIIt0370.015328IIIINNBN
2VSSA200672IVAt3240.0150.50.25IIAANNNN
3VSSA2006239IIIAt0370.015>6432IASynIBNBB
4VSSA200772IVAt1480.0150.50.25AAAANNNN
5VSSA2007239IIIt0370.0156416SynISynINNNN
6VSSA20075IIt0020.015>64>32IISynSynNNNN
7VSSA20075IIt0020.015>64>32IISynSynNNNN
8VSSA2007239IIIt0370.0156432SynSynSynSynNNBN
9VSSA20125IIt93530.015>6432IISynINNNN
10VSSA201272IVAt1480.0150.50.25IIIINNNN
11VSSA20125IIt24600.015328IAIINNNN
12VSSA201272IVAt3240.0150.250.25IIIINNNN
13VSSA20125IIt93530.0156432SynSynIIBNNN
14VSSA201372IVAt1480.0150.250.25IISynINBNN
15VSSA2013239IIIt138144IIIINNNN
16VISA2008239IIIAt03716168IISynINNBB
17VISA20095IIt2460166416AIIANNBN
18VISA201172IVAt3241610.25AIIIBBBB
19hVISA200672IVAt3240.0150.250.5IIIINNNN
20hVISA20065IIt0020.015168SynSynIINNNN
21hVISA20065IIt24600.015168SynISynSynNNNN
22hVISA2006239IIIt0370.015168SynSynSynSynNNNN
23hVISA20075IIt246016>6416IIIINNBN
24hVISA2007239IIIt0370.015328SynSynSynABBBN
25hVISA2007239IIIt0371688IIIINNBB
26hVISA20085IIt6010.015>6432ISynIINNNN
27hVISA201072IVAt1480.0150.50.5SynSynSynSynBBBB
28hVISA201172IVAt3240.0150.250.25IAIANNNN
29hVISA20115IIt93530.015328IISynINNNN
30hVISA20135IIt935316>6432IAIINNBB
VSSA, vancomycin-susceptible S. aureus; VISA, vancomycin-intermediate S. aureus; hVISA, heterogeneous VISA; ST, sequence type; SCC, staphylococcal cassette chromosome; MIC, minimum inhibitory concentration; R, rifampin; C, ciprofloxacin; L, levofloxacin; I, indifference; A, antagonism; Syn, synergy; B, bactericidal; N, non-bactericidal.
Table 3. Comparison of synergistic and antagonistic interactions of ciprofloxacin and levofloxacin combined with rifampin against methicillin-resistant Staphylococcus aureus strains.
Table 3. Comparison of synergistic and antagonistic interactions of ciprofloxacin and levofloxacin combined with rifampin against methicillin-resistant Staphylococcus aureus strains.
InteractionNumber of Bacterial Strains (%)
1× MIC0.5× MIC
Rifampin + CiprofloxacinRifampin + Levofloxacinp ValueRifampin + CiprofloxacinRifampin + Levofloxacinp Value
Synergy12
(43.3%)
6
(20.0%)
0.00828
(26.7%)
7
(23.3%)
0.5637
Antagonism2
(6.7%)
5
(16.7%)
0.08333
(10.0%)
5
(16.7%)
0.4142
MIC, minimum inhibitory concentration.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kang, Y.R.; Chung, D.R.; Ko, J.-H.; Huh, K.; Cho, S.Y.; Kang, C.-I.; Peck, K.R. Comparing the Synergistic and Antagonistic Interactions of Ciprofloxacin and Levofloxacin Combined with Rifampin against Drug-Resistant Staphylococcus aureus: A Time–Kill Assay. Antibiotics 2023, 12, 711. https://doi.org/10.3390/antibiotics12040711

AMA Style

Kang YR, Chung DR, Ko J-H, Huh K, Cho SY, Kang C-I, Peck KR. Comparing the Synergistic and Antagonistic Interactions of Ciprofloxacin and Levofloxacin Combined with Rifampin against Drug-Resistant Staphylococcus aureus: A Time–Kill Assay. Antibiotics. 2023; 12(4):711. https://doi.org/10.3390/antibiotics12040711

Chicago/Turabian Style

Kang, Yu Ri, Doo Ryeon Chung, Jae-Hoon Ko, Kyungmin Huh, Sun Young Cho, Cheol-In Kang, and Kyong Ran Peck. 2023. "Comparing the Synergistic and Antagonistic Interactions of Ciprofloxacin and Levofloxacin Combined with Rifampin against Drug-Resistant Staphylococcus aureus: A Time–Kill Assay" Antibiotics 12, no. 4: 711. https://doi.org/10.3390/antibiotics12040711

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