Next Article in Journal
Structure and Topology Prediction of Phage Adhesion Devices Using AlphaFold2: The Case of Two Oenococcus oeni Phages
Next Article in Special Issue
An Association of Pathogens and Biofilms with Alzheimer’s Disease
Previous Article in Journal
Predicting the Role of the Human Gut Microbiome in Constipation Using Machine-Learning Methods: A Meta-Analysis
Previous Article in Special Issue
Provisional Use of CLSI-Approved Quality Control Strains for Antimicrobial Susceptibility Testing of Mycoplasma (‘Mesomycoplasma’) hyorhinis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Synergism of Penicillin and Ceftriaxone against Enterococcus faecalis

1
Institute for Infectious Diseases and Infection Control, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany
2
Leibniz Center for Photonics in Infection Research, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany
3
Infectious Disease Unit, Auckland City Hospital, Auckland 1023, New Zealand
*
Author to whom correspondence should be addressed.
Submission received: 23 August 2021 / Revised: 7 October 2021 / Accepted: 10 October 2021 / Published: 14 October 2021
(This article belongs to the Special Issue Antimicrobial Testing (AMT))

Abstract

:
Enterococcus faecalis infective endocarditis is commonly treated with intravenous ampicillin/ceftriaxone combination therapy. Ampicillin, however, is unsuitable for outpatient parenteral antibiotic therapy (OPAT) regimens due to its instability in 24 h continuous infusors, and has been successfully replaced by benzylpenicillin used together with ceftriaxone in a few small case series. Since in vitro synergy data of penicillin/ceftriaxone against E. faecalis are still lacking, checkerboard assays were performed for 28 clinical E. faecalis isolates and one laboratory standard strain. Synergistic effects (both lowest and median FICI) were observed for penicillin/ceftriaxone in 15/29 isolates, while ampicillin/ceftriaxone exhibited synergism in 22/29 isolates. For isolates with ceftriaxone MICs ≤ 256 mg/L, the addition of free ceftriaxone trough concentrations to penicillin or ampicillin resulted in comparable synergistic effects for both combinations. In contrast, for isolates with ceftriaxone MICs ≥ 512 mg/L free ceftriaxone trough concentrations were only sufficient to exhibit synergistic effects in combination with ampicillin, but not penicillin. This study suggests that benzylpenicillin/ceftriaxone would be expected to be suitable for the OPAT treatment of enterococcal endocarditis for E. faecalis isolates with ceftriaxone MICs ≤ 256 mg/L. However, combination therapy would be expected to provide no advantage over benzylpenicillin monotherapy for isolates with ceftriaxone MICs ≥ 512 mg/L. Further investigation is required to analyse the relationship between ceftriaxone susceptibility and penicillin/ceftriaxone synergy, especially for isolates with ceftriaxone MICs of 64 to 512 mg/L.

1. Introduction

Enterococcus faecalis is an increasingly common cause of infective endocarditis (IE) and should be treated with prolonged synergistic, bactericidal antibiotic combination therapy [1,2]. Contemporary treatment guidelines recommend the use of intravenous (IV) ampicillin, amoxicillin, or benzylpenicillin combined with IV gentamicin for gentamicin susceptible isolates, or IV ampicillin combined with IV ceftriaxone for both high-level aminoglycoside-resistant (HLAR) and non-HLAR isolates [2,3]. Ceftriaxone is a more attractive synergy antibiotic than gentamicin, since it causes significantly fewer adverse effects [1,4].
Although a previous guideline for the Outpatient Parenteral Antibiotic Therapy (OPAT) treatment of IE was restrictive [5], a recent prospective cohort study has shown that the OPAT treatment of a broader range of patients with IE provided excellent results [6]. The abovementioned guideline-recommended regimens for the treatment of E. faecalis IE (EFIE) may be challenging to administer via an OPAT service due to the multiple doses required per day. The administration of the treatment by community nurses is often limited to once or twice daily injections or changing a 24 h elastomeric continuous infusor. While ampicillin/ceftriaxone has been proposed as an OPAT regimen for EFIE utilising either elastomeric continuous infusors or programmable pumps for the delivery of ampicillin [7], the literature shows conflicting results regarding the stability of ampicillin in elastomeric continuous infusors [8,9,10,11]. While ampicillin, which cannot be applied orally for reasons of bioavailability, could theoretically be replaced by orally administered amoxicillin, the clinical evidence supporting oral antibiotic regimens for treating EFIE is scarce. The use of oral antibiotics as part of the treatment of endocarditis has only been assessed in one trial [12] that enrolled a tightly defined group of patients with left-sided endocarditis who were transitioned to oral regimens. The patients with EFIE were treated with a variety of oral antibiotic combinations, the most common of which were amoxicillin /moxifloxacin, amoxicillin/linezolid, and amoxicillin/rifampicin. Oral amoxicillin/IV ceftriaxone has not been tested in a trial situation, as far as we are aware, and should therefore only be administered as part of a very carefully designed trial.
IV benzylpenicillin has been used as an alternative to IV ampicillin/amoxicillin for the treatment of EFIE, since benzylpenicillin is more stable in elastomeric continuous infusors and therefore better suited for OPAT regimens. The use of intravenous benzylpenicillin/ceftriaxone for the OPAT-guided treatment of E. faecalis endocarditis is common in New Zealand and also occurs in Australia and America. Benzylpenicillin has been successfully combined with ceftriaxone for the OPAT treatment of enterococcal endocarditis in a few small clinical case series [13,14,15], but in vitro synergy data supporting the use of benzylpenicillin with ceftriaxone for OPAT regimens are still lacking.
Here, we report the checkerboard synergy analysis of penicillin/ceftriaxone and ampicillin/ceftriaxone in 28 clinical E. faecalis isolates and one laboratory standard strain, allowing for a comparison of both combinations at concentrations achieved by OPAT dosage regimens.

2. Materials and Methods

2.1. Enterococcal Strains and Antibiotics

Clinical E. faecalis isolates were obtained from blood cultures from, or the heart valves of, patients with enterococcal infection at various foci, including eight patients with infective endocarditis (Table 1). The antibiotic-susceptibility data for all clinical isolates are depicted in Table S1. All patients were admitted to Jena University Hospital, Germany, in 2015. E. faecalis ATCC 29,212 served as a reference strain. Bacterial liquid cultures were prepared in Todd Hewitt broth (Karl Roth, Karlsruhe, Germany). Test solutions of ampicillin (AMP) (Karl Roth, Karlsruhe, Germany), ceftriaxone (CRO) (TCI Europe, Zwijndrecht, Belgium), and benzylpenicillin (PEN) (InfectoPharm, Heppenheim, Germany) were prepared immediately before use.

2.2. Synergism Testing by Checkerboard Assays

Checkerboard assays for penicillin/ceftriaxone were performed as described previously [17], with 11 and 7 serial 2-fold dilution steps for benzylpenicillin and ceftriaxone, respectively. The benzylpenicillin concentrations tested were chosen to cover the penicillin breakpoint range. The ceftriaxone concentrations tested included those that approximate the range of estimated or measured mean free plasma ceftriaxone trough concentrations expected to be achieved with OPAT ceftriaxone-dosing regimens of 2 g every 12 h, 4 g once daily, or 2 g once daily (4, 1.5, and 1 mg/L, respectively) [18,19,20], hereafter referred to as “free ceftriaxone trough concentrations”. The checkerboard assays used for ampicillin/ceftriaxone were partly assessed in a previous study [21]; however, they were newly evaluated according to novel EUCAST guidelines [16] (see Table 1, footnote c), which has led to a change in MICs and fractional inhibitory concentrations indices (FICI) for 10 out of 21 previously tested isolates. Checkerboards for ampicillin/ceftriaxone were repeated for eight of these isolates to guarantee the reproducibility of previous results. Each checkerboard assay was performed in duplicate. The effects of the combined antibiotics were evaluated by calculating the FICI along the turbidity/non-turbidity interface using the following formula:
FICI penicillin / ceftriaxone = MIC   penicillin   ( combination ) MIC   penicillin   ( alone ) + MIC   ceftriaxone   ( combination ) MIC   ceftriaxone   ( alone ) .
Three different methods for interpreting the FICIs were used: (i) the lowest FICI value, with values ≤ 0.5 indicating synergism and 0.5 < FICI < 4 indicating no interaction; (ii) the median FICI value, taking 0.8 as the synergy threshold; (iii) single, one-point FICIs at free ceftriaxone trough concentrations.

2.3. Statistical Analysis

The correlation of the MICs and FICIs was tested using the nonparametric Spearman’s rank-correlation coefficient (rs) with a two-tailed CI of 95%.

3. Results

Synergistic effects were observed for penicillin/ceftriaxone in 16 (lowest FICI method) and 21 (median FICI method) of the 29 tested E. faecalis strains (Table 1). Ampicillin/ceftriaxone synergised in 22 strains (both lowest and median FICI method) (Table 1). Ampicillin MICs (MICAMP) ranged from 0.25 to 2 mg/L, while penicillin MICs (MICPEN) ranged from 0.5 to 4 mg/L, the highest MICPEN of 4 mg/L being below the CLSI susceptibility breakpoint of 8 mg/L. Ceftriaxone MICs (MICCRO) ranged from 1–2 to more than 1024 mg/L. The MICAMP showed no correlation with the MICCRO, but the MICPEN exhibited a weak positive correlation with the MICCRO (rs = 0.52, p = 0.004). Both the MICPEN and MICCRO displayed a strong inverse correlation with the FICI values of penicillin/ceftriaxone (FICIPEN/CRO)—i.e., the higher the MICPEN or MICCRO, the lower the FICIPEN/CRO (MICPEN rs = −0.61, p = 0.001; MICCRO rs = −0.71, p < 0.001). The MICCRO inversely correlated with the FICIAMP/CRO (rs = −0.76, p < 0.001), but the MICAMP did not.
To analyse whether the synergistic effects determined by the lowest and median FICI methods were achieved at free ceftriaxone trough concentrations, the lowest sub-MIC ceftriaxone concentrations that resulted in a reduction in the MICPEN or MICAMP to one half, one quarter, and one eighth of the MICPEN or MICAMP are recorded in Table 2, together with the corresponding single, one-point FICIs. For 13 isolates (9367; 1653; 848; 5597; 6886; 67,230; 2164; 4497; 11,223; 22,424; 10,485; 13,703; 7914), synergy between penicillin/ceftriaxone occurred at free ceftriaxone trough concentrations, while for another 8 isolates (5187; 245; ATCC; 6747; 8669; 281; 26,786; 404), the addition of these free ceftriaxone trough concentrations resulted in a four-fold reduction in the MICPEN but with FICIs above the synergy threshold of 0.5. In contrast, ampicillin/ceftriaxone exhibited synergism in 23 isolates at free ceftriaxone trough concentrations. For the isolates with MICCRO ≥ 512 mg/L, lower sub-MIC ceftriaxone concentrations were required in combination with ampicillin than in combination with penicillin to produce the same effect (Table 2). Here, the addition of free ceftriaxone trough concentrations to penicillin resulted in a reduction in the MICPEN in one of seven isolates, whereas the addition to ampicillin reduced the MICAMP in six of seven isolates.

4. Discussion

The synergy analysis of ampicillin/ceftriaxone and penicillin/ceftriaxone in 29 E. faecalis isolates revealed that both combinations have comparable synergistic effects for isolates with MICCRO ≤ 256 mg/L. A reduction in the MICPEN with the addition of ceftriaxone concentrations approximating free ceftriaxone trough concentrations was shown in all of these isolates. In contrast, for isolates with MICCRO ≥ 512 mg/L discordance was seen between the synergy provided by ampicillin/ceftriaxone and by penicillin/ceftriaxone at free ceftriaxone trough concentrations. These ceftriaxone concentrations were insufficient in combination with penicillin, but sufficient in combination with ampicillin to produce a synergistic effect for isolates with an MICCRO ≥ 512 mg/L.
While a synergistic effect between ampicillin and ceftriaxone in E. faecalis is not a new finding, and in fact forms part of the basis for recommending this combination for the treatment of EFIE, there are currently no data available showing a potential synergistic effect between penicillin and ceftriaxone. Importantly, our study shows that a similar synergistic effect between penicillin and ceftriaxone cannot be assumed just because there is a proven synergistic effect between ampicillin and ceftriaxone. Although our sample size is relatively small, with 29 E. faecalis isolates, the study on which the current guidelines for the use of ampicillin and ceftriaxone for the treatment of EFIE is based contained just 10 isolates [22]. Our data show that the suitability of penicillin/ceftriaxone for the treatment of EFIE likely depends on the specific isolate’s ceftriaxone susceptibility. However, the interpretation of the relationship between ceftriaxone susceptibility and penicillin/ceftriaxone synergy in this study is limited by the lack of isolates with an MICCRO of 64 and 128 mg/L, as well as by the limited number of isolates with an MICCRO of 256 and 512 mg/L. Our E. faecalis strain collection comprises 50 clinical isolates collected between 2015 and 2017. The ceftriaxone MIC testing of this collection shows two populations: a larger population of isolates (68%) centred around 8 mg/L ± 1–2× MIC and a smaller population of isolates (32%) centered around 512 mg/L ± 1× MIC (Figure S1). None of our isolates exhibited a ceftriaxone MIC of 64 or 128 mg/L. The EUCAST MICCRO distribution data for E. faecalis show that, of the 8314 submitted clinical isolates, only 5% had an MICCRO ≤ 32 mg/L [23], which contrasts strongly with the MICCRO distribution of our cohort.
Caution should be applied when drawing conclusions from microbiological studies for use in the clinical setting. While the checkerboard assay has been intensively used for studying antibiotic interactions, clinical correlation studies linking in vitro synergy data to direct treatment outcomes are lacking. The methodology of the checkerboard assay does have some limitations, such as a high degree of variability in the selection of the wells used for the final FICI calculation [24,25]. To compensate for a potential selection bias leading to an overestimation of the synergistic effect, we used the median FICI, the lowest FICI, and one-point FICIs at free ceftriaxone trough concentrations for a clinically meaningful interpretation of the checkerboard assay.
The synergistic effect of dual beta-lactam therapy is thought to be based on the complementary inhibition of penicillin binding protein (PBP) homologues, resulting in the inhibition of cell-wall synthesis. Little is known about the detailed functions of PBPs in E. faecalis, and this understanding is complicated by the inconsistent labelling of the different PBP homologues [26]. The synergism of amoxicillin/cefotaxime in a single E. faecalis isolate was postulated to be explained by the partial saturation of the essential PBPs 4 and 5 by amoxicillin, coupled with the complete saturation of the non-essential PBPs 2 and 3 by very low cefotaxime concentrations [27]. Ceftriaxone resistance is known to be mediated by the overproduction and mutations of PBPs 4 and 5 as well as other, novel low-affinity class-B PBPs, further reducing the already low affinity of ceftriaxone for these essential PBPs [26]. The discordance of the synergistic effects between ampicillin/ceftriaxone and penicillin/ceftriaxone in isolates with high MICCRO might be explained by the more complete saturation of the ceftriaxone-resistance-mediating PBP profile by ampicillin than by penicillin. The difference in saturation would mean that higher ceftriaxone concentrations would be required to compensate for the poorer binding of penicillin to the altered PBPs. This explanation is supported by the fact that the MICPEN, but not the MICAMP, positively correlated with the MICCRO, indicating that both antibiotics target similar PBP homologues. Interestingly, a study with one E. faecalis isolate showed no interaction between penicillin/ceftriaxone, but synergistic interaction between penicillin/ceftaroline, which is a novel cephalosporin with enhanced affinity to PBP 5 [28]. This further supports the hypothesis that the incomplete binding of PBP 5 by penicillin and ceftriaxone counteracts the synergistic effect.
The addition of ceftriaxone to benzylpenicillin provides synergy, or at least partial synergy, for E. faecalis isolates with an MICCRO ≤ 256 mg/L. No significant benefit from adding ceftriaxone to benzylpenicillin is expected for any isolate with an MICCRO ≥ 512 mg/L. These microbiological data support the use of OPAT treatment with the continuous infusion of benzylpenicillin and ceftriaxone for EFIE for isolates with an MICCRO ≤ 256 mg/L. As the penicillin/ceftriaxone synergy-testing data for isolates with MICCRO of 64 to 512 mg/L are limited in this study, further investigation is required to establish a reliable MICCRO cut-off above which the combination of benzylpenicillin and ceftriaxone is not superior to benzylpenicillin alone.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/microorganisms9102150/s1: Figure S1: Ceftriaxone MIC distribution data for the E. faecalis strain collection; Table S1: Antibiotic susceptibility results determined by Vitek 2 (bioMerieux, Craponne, France).

Author Contributions

Conceptualisation, L.T., S.B., and E.D.; methodology, L.T., S.B., and E.D.; validation, L.T., S.B., and E.D.; formal analysis, L.T.; investigation, L.T.; resources, L.T.; data curation, L.T.; writing—original draft preparation, L.T.; writing—review and editing, L.T., S.B., E.D., O.M., and M.W.P.; visualisation, L.T.; supervision, M.W.P.; project administration, L.T.; funding acquisition, O.M., M.W.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Federal Ministry of Education and Research, Germany (grant numbers 01KI1501 and 13N15467).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No further datasets are available.

Acknowledgments

The graphical abstract was created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fernández-Hidalgo, N.; Escolà-Vergé, L.; Pericàs, J.M. Enterococcus faecalis endocarditis: What’s next? Future Microbiol. 2020, 15, 349–364. [Google Scholar] [CrossRef]
  2. Habib, G.; Lancellotti, P.; Antunes, M.J.; Bongiorni, M.G.; Casalta, J.P.; Del Zotti, F.; Dulgheru, R.; El Khoury, G.; Erba, P.A.; Iung, B.; et al. ESC Guidelines for the management of infective endocarditis: The task force for the management of infective endocarditis of the European Society of Cardiology (ESC). Endorsed by: European Association for Cardio-Thoracic Surgery (EACTS), the European Association of Nuclear Medicine (EANM). Eur. Heart J. 2015, 36, 3075–3128. [Google Scholar] [PubMed]
  3. Baddour, L.M.; Wilson, W.R.; Bayer, A.S.; Fowler, V.G., Jr.; Tleyjeh, I.M.; Rybak, M.J.; Barsic, B.; Lockhart, P.B.; Gewitz, M.H.; Levison, M.E.; et al. Infective endocarditis in adults: Diagnosis, antimicrobial therapy, and management of complications: A scientific statement for healthcare professionals from the american heart association. Circulation 2015, 132, 1435–1486. [Google Scholar] [CrossRef] [PubMed]
  4. El Rafei, A.; DeSimone, D.C.; Narichania, A.D.; Sohail, M.R.; Vikram, H.R.; Li, Z.; Steckelberg, J.M.; Wilson, W.R.; Baddour, L.M. Comparison of Dual β-Lactam therapy to penicillin-aminoglycoside combination in treatment of Enterococcus faecalis infective endocarditis. J. Infect. 2018, 77, 398–404. [Google Scholar] [CrossRef] [PubMed]
  5. Andrews, M.-M.; von Reyn, C.F. Patient selection criteria and management guidelines for outpatient parenteral antibiotic therapy for native valve infective endocarditis. Clin. Infect. Dis. 2001, 33, 203–209. [Google Scholar] [CrossRef] [Green Version]
  6. Pericà, S.J.M.; Llopis, J.; González-Ramallo, V.; Goenaga, M.Á.; Muñoz, P.; García-Leoni, M.E.; Fariñas, M.C.; Fariñas, M.C.; Ambrosioni, J.; Luque, R.; et al. Outpatient parenteral antibiotic treatment for infective endocarditis: A prospective cohort study from the GAMES cohort. Clin. Infect. Dis. 2019, 69, 1690–1700. [Google Scholar] [CrossRef] [PubMed]
  7. Gil-Navarro, M.V.; Lopez-Cortes, L.E.; Luque-Marquez, R.; Galvez-Acebal, J.; de Alarcon-Gonzalez, A. Outpatient parenteral antimicrobial therapy in Enterococcus faecalis infective endocarditis. J. Clin. Pharm. Ther. 2018, 43, 220–223. [Google Scholar] [CrossRef]
  8. Nakamura, T.; Enoki, Y.; Uno, S.; Uwamino, Y.; Iketani, O.; Hasegawa, N.; Matsumoto, K. Stability of benzylpenicillin potassium and ampicillin in an elastomeric infusion pump. J. Infect. Chemother. 2018, 24, 856–859. [Google Scholar] [CrossRef]
  9. Tice, A.D.; Rehm, S.J.; Dalovisio, J.R.; Bradley, J.S.; Martinelli, L.P.; Graham, D.R.; Gainer, R.B.; Kunkel, M.J.; Yancey, R.W.; Williams, D.N.; et al. Practice guidelines for outpatient parenteral antimicrobial therapy. Clin. Infect. Dis. 2004, 38, 1651–1671. [Google Scholar] [CrossRef] [Green Version]
  10. Maher, M.; Jensen, K.J.; Lee, D.; E Nix, D. Stability of ampicillin in normal saline and buffered normal saline. Int. J. Pharm. Compd. 2016, 20, 338–342. [Google Scholar]
  11. Huskey, M.; Lewis, P.; Brown, S.D. Stability of ampicillin in normal saline following refrigerated storage and 24-hour pump recirculation. Hosp. Pharm. 2020. [Google Scholar] [CrossRef]
  12. Iversen, K.; Ihlemann, N.; Gill, S.U.; Madsen, T.; Elming, H.; Jensen, K.T.; Bruun, N.E.; Høfsten, D.E.; Fursted, K.; Christensen, J.J.; et al. Partial oral versus intravenous antibiotic treatment of endocarditis. N. Engl. J. Med. 2019, 380, 415–424. [Google Scholar] [CrossRef] [PubMed]
  13. Suzuki, H.; Carlson, J.R.; Matsumoto, E. Treatment of enterococcus faecalis infective endocarditis with penicillin G plus ceftriaxone. J. Infect. Dis. 2020, 52, 135–138. [Google Scholar] [CrossRef]
  14. Tritle, B.J.; Timbrook, T.T.; Fisher, M.A.; Spivak, E.S. Penicillin as a potential agent for dual β-lactam therapy for enterococcal endocarditis. Clin Infect Dis. 2020, 70, 1263–1264. [Google Scholar] [CrossRef] [PubMed]
  15. Briggs, S.; Broom, M.; Duffy, E.; Everts, R.; Everts, G.; Lowe, B.; McBride, S.; Bhally, H. Outpatient continuous-infusion benzylpenicillin combined with either gentamicin or ceftriaxone for enterococcal endocarditis. J. Antimicrob. Chemother. 2021. [Google Scholar] [CrossRef] [PubMed]
  16. EUCAST. EUCAST Reading Guide for Broth Microdilution, Version 3.0 January 2021. Available online: https://www.eucast.org/ast_of_bacteria/mic_determination/?no_cache=1 (accessed on 5 March 2021).
  17. Stein, C.; Makarewicz, O.; Bohnert, J.A.; Pfeifer, Y.; Kesselmeier, M.; Hagel, S.; Pletz, M.W. Three dimensional checkerboard synergy analysis of colistin, meropenem, tigecycline against multidrug-resistant clinical klebsiella pneumonia isolates. PLoS ONE 2015, 10, e0126479. [Google Scholar] [CrossRef] [Green Version]
  18. Herrera-Hidalgo, L.; de Alarcón, A.; López-Cortes, L.E.; Luque-Márquez, R.; López-Cortes, L.F.; Gutiérrez-Valencia, A.; Gil-Navarro, M.V. Is once-daily high-dose ceftriaxone plus ampicillin an alternative for enterococcus faecalis infective endocarditis in outpatient parenteral antibiotic therapy programs? Antimicrob. Agents Chemother. 2020, 65, e02099-20. [Google Scholar] [CrossRef] [PubMed]
  19. Patel, I.H.; Chen, S.; Parsonnet, M.; Hackman, M.R.; A Brooks, M.; Konikoff, J.; A Kaplan, S. Pharmacokinetics of ceftriaxone in humans. Antimicrob. Agents Chemother. 1981, 20, 634–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Pollock, A.A.; Tee, P.E.; Patel, I.H.; Spicehandler, J.; Simberkoff, M.S.; Rahal, J.J. Pharmacokinetic characteristics of intravenous ceftriaxone in normal adults. Antimicrob. Agents Chemother. 1982, 22, 816–823. [Google Scholar] [CrossRef] [Green Version]
  21. Thieme, L.; Klinger-Strobel, M.; Hartung, A.; Stein, C.; Makarewicz, O.; Pletz, M.W. In vitro synergism and anti-biofilm activity of ampicillin, gentamicin, ceftaroline and ceftriaxone against Enterococcus faecalis. J. Antimicrob. Chemother. 2018, 73, 1553–1561. [Google Scholar] [CrossRef] [Green Version]
  22. Gavaldà, J.; Torres, C.; Tenorio, C.; López, P.; Zaragoza, M.; Capdevila, J.A.; Almirante, B.; Ruiz, F.; Borrell, N.; Gomis, X.; et al. Efficacy of ampicillin plus ceftriaxone in treatment of experimental endocarditis due to enterococcus faecalis strains highly resistant to aminoglycosides. Antimicrob. Agents Chemother. 1999, 43, 639–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. EUCAST. EUCAST MIC Distributions for Ceftriaxone. Available online: https://mic.eucast.org/search/?search%5Bmethod%5D=mic&search%5Bantibiotic%5D=57&search%5Bspecies%5D=-1&search%5Bdisk_content%5D=-1&search%5Blimit%5D=50 (accessed on 5 March 2021).
  24. Odds, F.C. Synergy, antagonism, and what the chequerboard puts between them. J. Antimicrob. Chemother. 2003, 52, 1. [Google Scholar] [CrossRef] [PubMed]
  25. Bonapace, C.R.; Bosso, J.A.; Friedrich, L.V.; White, R.L. Comparison of methods of interpretation of checkerboard synergy testing. Diagn. Microbiol. Infect. Dis. 2002, 44, 363–366. [Google Scholar] [CrossRef]
  26. Djorić, D.; Little, J.L.; Kristich, C.J. Multiple low-reactivity class b penicillin-binding proteins are required for cephalosporin resistance in enterococci. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef]
  27. Mainardi, J.L.; Gutmann, L.; Acar, J.F.; Goldstein, F.W. Synergistic effect of amoxicillin and cefotaxime against Enterococcus faecalis. Antimicrob. Agents Chemother. 1995, 39, 1984–1987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Cusumano, J.; Daffinee, K.; Bodo, E.; Laplante, K. Alternative Dual Beta-Lactam Combinations for Enterococcus Faecalis Infective Endocarditis; abstract P2804; ECCMID: Amsterdam, The Netherlands, 2019. [Google Scholar]
Table 1. Susceptibility (MIC) and synergy (FICI) results for penicillin/ceftriaxone versus ampicillin/ceftriaxone in the patient cohort.
Table 1. Susceptibility (MIC) and synergy (FICI) results for penicillin/ceftriaxone versus ampicillin/ceftriaxone in the patient cohort.
Isolate aClinical Background bGenderAge (Years)MIC CRO (mg/L)MIC PEN (mg/L)MIC AMP (mg/L)FICI d of PEN/CROFICI of AMP/CRO
5187urosepsismale791–2 c110.75 (1.13)0.63 (0.88)
245endocarditismale762 c10.50.75 (1.03)0.63 (0.88)
26,786endocarditismale67210.250.56 (0.75)0.74 (1.05)
404endocarditisfemale7840.50.250.75 (0.75)0.49 (0.68)
ATCC///4110.75 (0.75)0.50 (0.69)
6747biliary tract infectionmale814 c110.75 (0.75)0.50 (0.63)
11,223endocarditisfemale65820.50.38 (0.56)0.31 (0.50)
22,424endocarditismale68810.50.25 (0.56)0.37 (0.47)
8669OImale808 c110.63 (0.75)0.25 (0.38)
9367recurrent bacteraemiafemale858 c220.50 (0.63)0.38 (0.63)
1653urosepsisfemale878 c210.38 (0.63)0.38 (0.56)
848OImale608210.38 (0.56)0.31 (0.47)
5597OIfemale558–16 c210.31 (0.52)0.50 (0.63)
10,485endocarditismale741610.50.38 (0.81)0.24 (0.53)
6886wound infectionfemale6816 c120.63 (0.75)0.31 (0.52)
281sepsisfemale7916 c110.50 (0.63)0.31 (0.38)
67,230endocarditismale3916210.38 (0.47)0.25 (0.45)
2164OIfemale7816 c220.25 (0.56)0.38 (0.63)
4497urosepsisfemale6732 c210.31 (0.45)0.38 (0.56)
10,021urosepsisfemale423210.50.56 (1.01)0.15 (0.34)
13,703bacteraemiamale54256210.16 (0.27)0.16 (0.37)
7914OImale5425610.250.25 (0.38)0.26 (0.37)
905endocarditismale75512210.31 (0.52)0.27 (0.44)
6037urosepsismale861024 c110.27 (0.51)0.12 (0.25)
7183urosepsismale761024210.31 (0.55)0.38 (0.52)
3043OIfemale74>102420.5N. A.N. A.
8653OImale56>102421N. A.N. A.
3062urosepsismale77>102441N. A.N. A.
9190wound infectionfemale59>102441N. A.N. A.
a E. faecalis isolates were obtained from the Institute of Medical Microbiology in Jena, Germany. All clinical isolates originated from blood cultures except for 67230 and 245, which were sampled by swabs from infected mitral valves. / = N.A. to the laboratory standard strain ATCC 29,212. b OI = opportunistic infection. c In some isolates, ceftriaxone treatment led to trailing MIC endpoints, with wells showing the same level of turbidity observed in the growth control; followed by wells with less, but still visible, turbidity; and eventually wells with pinpoint growth (small aggregates). According to the EUCAST reading guide for broth microdilution [16], pinpoint growth was disregarded and recorded as the MIC. d FICI values are given as the lowest FICI with the median FICI in parentheses. Synergistic FICI values (lowest FICI ≤ 0.5 and median FICI ≤ 0.8) are indicated in bold. N.A. = not determined due to MIC > 1024 mg/L.
Table 2. Lowest sub-MIC CRO concentrations resulting in two-, four-, and eight-fold reductions in the effective MICPEN or MICAMP and corresponding FICI values of the resulting concentration combinations.
Table 2. Lowest sub-MIC CRO concentrations resulting in two-, four-, and eight-fold reductions in the effective MICPEN or MICAMP and corresponding FICI values of the resulting concentration combinations.
IsolateMICCRO Alone [mg/L]Lowest CRO Concentration Resulting in Two-Fold Reduction in MICPEN or MICAMPLowest CRO Concentration Resulting in Four-Fold Reduction in MICPEN or MICAMPLowest CRO Concentration Resulting in Eight-Fold Reduction in MICPEN or MICAMP
PenicillinAmpicillinPenicillinAmpicillinPenicillinAmpicillin
CRO conc. [mg/L]FICI aCRO conc. [mg/L]FICICRO conc. [mg/L]FICICRO conc. [mg/L]FICICRO conc. [mg/L]FICICRO conc. [mg/L]FICI
51871–211.500.51.0011.250.50.75N.A.N.A.0.50.625
24520.50.750.50.7510.750.50.50N.A.N.A.0.50.375
26,78620.250.6250.50.7510.7510.7410.625N.A.N.A.
404410.7510.7520.7510.49N.A.N.A.20.625
ATCC410.750.50.62520.7510.50N.A.N.A.20.625
6747410.750.50.62520.7510.50N.A.N.A.20.625
11,223810.62510.62510.37510.37540.62540.625
22,424810.62510.62520.510.37540.62520.375
86698 10.6250.50.56340.7510.375N.A.N.A.20.375
9367810.6250.50.56320.501–20.5040.62520.375
1653810.6250.50.5631–20.5010.37540.6252–40.625
848810.6250.5–10.62510.37510.37540.62520.375
55978–1610.6250.50.56320.5020.2581.12540.625
10,4851610.560.250.5220.37510.3140.37520.25
68861610.5630.5–10.56340.5010.31340.37540.375
281161–20.6250.50.534–80.7510.31380.62540.375
67,2301610.5630.5–10.56320.3751–20.37540.37540.375
21641610.56310.56320.3752–40.5040.37580.625
44973210.530.50.5632–40.37520.31380.37540.25
10,0213220.560.1250.50N.A.N.A.0.50.266N.A.N.A.20.18
13,70325610.5010.5010.2610.25160.2580.16
791425610.5010.5080.2840.26640.375640.375
9055122–40.5110.50320.3134–80.266N.A.N.A.640.25
6037102480.510.50.501280.3750.5–10.2515120.6252–40.13
71831024160.520.5–10.50640.313640.3132560.375N.A.N.A.
3043>1024512N.A.4N.A.1024N.A.128N.A.N.A.N.A.N.A.N.A.
8653>1024512N.A.2N.A.N.A.N.A.N.A.N.A.N.A.N.A.N.A.N.A.
3062>1024128N.A.32–64N.A.256–512N.A.256N.A.512–1024N.A.N.A.N.A.
9190>102464–128N.A.2N.A.512N.A.256N.A.N.A.N.A.N.A.N.A.
a Synergistic FICI values ≤ 0.5 obtained at free ceftriaxone trough concentrations (CRO conc. = 1 to 4 mg/L) are indicated in bold. Two-, four- and eight-fold reductions in MICPEN or MICAMP correspond to fractional inhibitory concentration (FIC) values (FICA = MICA combined/MICA alone; FICA + FICB = FICIA/B) of 0.5, 0.25 and 0.125. The respective FICs of ceftriaxone are calculated by dividing the lowest sub-MIC CRO concentration (CRO conc.) by the MICCRO. N.A. = the respective reduction in MICPEN or MICAMP not reached.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Thieme, L.; Briggs, S.; Duffy, E.; Makarewicz, O.; Pletz, M.W. In Vitro Synergism of Penicillin and Ceftriaxone against Enterococcus faecalis. Microorganisms 2021, 9, 2150. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9102150

AMA Style

Thieme L, Briggs S, Duffy E, Makarewicz O, Pletz MW. In Vitro Synergism of Penicillin and Ceftriaxone against Enterococcus faecalis. Microorganisms. 2021; 9(10):2150. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9102150

Chicago/Turabian Style

Thieme, Lara, Simon Briggs, Eamon Duffy, Oliwia Makarewicz, and Mathias W. Pletz. 2021. "In Vitro Synergism of Penicillin and Ceftriaxone against Enterococcus faecalis" Microorganisms 9, no. 10: 2150. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9102150

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