Skip to main content
PLOS One logoLink to PLOS One
. 2025 Sep 5;20(9):e0331440. doi: 10.1371/journal.pone.0331440

In Vitro synergy of Farnesyltransferase inhibitors in combination with colistin against ESKAPE bacteria

Marian Klose 1, Lea Weber 1, Hagen Sjard Bachmann 1,*
Editor: Shengwei Sun,2
PMCID: PMC12412981  PMID: 40911556

Abstract

The emergence of antibiotic resistance continues to pose a significant global challenge. Drug repurposing, wherein existing therapeutics are evaluated for new applications, offers a promising strategy to address this issue. Farnesyltransferase inhibitors (FTIs), initially developed for cancer therapy, have demonstrated antimicrobial activity against several gram-positive bacteria. This study investigates their activity in combination with colistin against gram-positive and gram-negative bacteria. We focus on key ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) pathogens while incorporating additional bacterial strains to provide a comprehensive understanding of differential responses and potential dose-dependent synergistic effects. Antimicrobial susceptibility was assessed using broth microdilution, while synergy was evaluated through checkerboard, time-kill, and growth kinetics assays. When combined with sub-inhibitory colistin, FTIs inhibited gram-negative bacterial growth. Tipifarnib exhibited more potent antimicrobial activity against gram-negative strains than lonafarnib. Peptidomimetic FTIs, B581 and FTI-277, inhibited gram-negative bacteria in combination with colistin but had no effect on the gram-positive strains tested. In contrast, alpha-hydroxy farnesyl phosphonic acid, an FPP analog, and bempedoic acid, targeting the mevalonate pathway, showed no antibacterial activity. In addition to their known inhibition of gram-positive bacteria, FTIs exhibited efficacy against gram-negative bacteria, including colistin-resistant Enterobacter cloacae subsp. cloacae, when combined with sub-inhibitory colistin. This might be due to a mechanism distinct from their eukaryotic targets, potentially involving the disruption of multiple biosynthetic pathways. Future studies will focus on elucidating these mechanisms of FTIs and exploring the therapeutic potential of FTI/colistin combinations against ESKAPE and other multidrug-resistant pathogens.

Importance

Antibiotic resistance is a growing threat to global health, necessitating innovative strategies to combat resistant pathogens. This study highlights the potential of farnesyltransferase inhibitors (FTIs), originally developed for cancer therapy, as antimicrobials against key pathogenic bacteria. When combined with colistin, FTIs demonstrated significant activity against gram-negative bacteria. Notably, this combination enabled FTIs to exert antimicrobial activity even against colistin-resistant bacteria. Although the exact mechanism of action remains to be elucidated, this combination may offer a valuable therapeutic option against bacteria that have developed increased resistance to colistin, addressing a critical need in the treatment of multidrug-resistant pathogens.

Introduction

The emergence of antibiotic resistance is a steadily growing concern [1]. At the same time, research into novel antimicrobial agents is proving difficult and cost-intensive [2]. If current trends continue, it is estimated that antibiotic-resistant bacteria alone could cause over 10 million deaths per year by 2050 [3]. One way to at least partially address these problems is to test already established drugs of other medical areas for possible antimicrobial effects (drug repurposing). This was successfully conducted by using statins, which are lipid-modifying agents due to their competitive binding to 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. One of the multiple pleiotropic properties also revealed antimicrobial potential in addition to its main effect [4].

Likewise, we were able to show that inhibitors of farnesyltransferase (FTase), which are used as cancer therapeutics, also exhibit antimicrobial effects [5]. However, antimicrobial effects of farnesyltransferase inhibitors (FTIs) have so far only been demonstrated in gram-positive bacteria (Fig 1A). Given that the outer membrane (OM) may limit the activity of FTIs, colistin, which binds to Lipid A in the lipopolysaccharide layer, thereby disrupting the OM’s integrity and increasing its permeability, could serve as a means to expand their spectrum of activity to gram-negative bacteria [8,9].

Fig 1. Antimicrobial activity of farnesyltransferase inhibitors. (A) shortly summarizes the antimicrobial activity of the farnesyltransferase inhibitors (FTIs) lonafarnib and tipifarnib, which demonstrated efficacy against gram-positive bacteria, including pathogenic Staphylococcus species. However, no activity was observed against gram-negative bacteria [5]. (B) gives an overview of the used drugs and their corresponding targets in eukaryotic cells. Drugs that show antimicrobial potential are marked (*): Statins, Bisphosphonates [6,7], and FTIs. (C) highlights the primary objective of this study: expanding the spectrum of FTIs to gram-negative bacteria by combining them with sub-inhibitory concentrations of colistin. This approach enables FTIs to exhibit antimicrobial activity against several gram-negative bacteria when co-administered with sub-inhibitory concentrations of colistin, while also potentially reducing the colistin dose required for growth inhibition, offering a novel strategy to combat antimicrobial resistance. Abbreviations: ACL, ATP-citrate lyase; AST, antimicrobial susceptibility testing; FPP, farnesyl pyrophosphate; FPPS, farnesyl pyrophosphate synthase; FTase, farnesyltransferase; GGPP, geranylgeranyl pyrophosphate; GPP, geranyl pyrophosphate; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; IPP, isopentenyl pyrophosphate; PP, pyrophosphate. Created in BioRender.

Fig 1

Similar studies have already been conducted with other drugs. For example, Huang, Zhu [10] were able to show that increased intracellular accumulation of flavomycin can be achieved with simultaneous administration of sub-inhibitory colistin. This led to a significant reduction in the amount of colistin required to reduce the bacterial cell count. While these findings highlight the potential of combination therapies, the antimicrobial effects of FTIs have not been broadly explored across all ESKAPE pathogens in a combinational approach with colistin. The ESKAPE panel, consisting of Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species, represents a major global health threat due to their ability to “escape” the effects of many current antibiotics [11,12].These pathogens are not only responsible for a significant proportion of nosocomial infections but also exhibit a high prevalence of multidrug resistance, complicating treatment options [13]. Their virulence and adaptive mechanisms enable rapid genomic adaptation and resistance development, making it even more critical to discover new treatment options [14,15].

In addition to pathogenic bacteria, interactions with commensal bacterial species are relevant when assessing novel antimicrobial approaches. The balance of microbial communities, such as those in the gut or on the skin, plays a critical role in maintaining host physiology [16]. Disrupting this balance through antimicrobial interventions could potentially promote the overgrowth of opportunistic pathogens [17], highlighting the importance of understanding antimicrobial effects beyond traditional pathogenic targets.

The specific objectives of this study are as follows: (i) to evaluate the effects of the non-peptidomimetic FTIs, lonafarnib and tipifarnib, on gram-positive and gram-negative bacteria when co-administered with colistin at sub-inhibitory concentrations; (ii) to assess the effects of the peptidomimetic FTIs, B581 and FTI-277, under similar conditions; and (iii) to investigate the impacts of alpha-hydroxy farnesyl phosphonic acid, a non-hydrolyzable analog of FPP, and (iv) bempedoic acid, an adenosine triphosphate citrate lyase inhibitor, under the same conditions, to better contextualize the effects of the aforementioned substances on bacterial behavior. Unlike the previously mentioned substances, which were primarily developed for the treatment of carcinomas, bempedoic acid is a lipid-lowering agent prescribed to a broader patient population. Notably, bempedoic acid and lonafarnib share the advantage of already being approved for clinical use [18,19], with bempedoic acid targeting the cholesterol biosynthesis pathway upstream of statins. Fig 1B provides an overview of these substances and their established eukaryotic targets. To achieve these aims, the checkerboard assay was employed to determine potential synergistic interactions. In addition, we extended the current checkerboard method by incorporating the viability agent resazurin and analyzing the results using fluorescence measurement. Moreover, dose-dependent interactions were systematically assessed, as illustrated in Fig 1C.

Materials and methods

Chemicals

Colistin was purchased as colistin sulfate from Sigma-Aldrich (Sigma Aldrich, MO, USA). All additional reference antibiotics were purchased from Roth (Carl Roth, Karlsruhe, Germany). Bempedoic acid (ETC-1002, abbreviated as BPD in this study), FTI-277 (as FTI-277 HCl), lonafarnib and tipifarnib were all purchased from SelleckChem (SelleckChem, Munich, Germany). B581 was purchased from Enzo Life Sciences (Enzo Biochem, Inc., NY, USA), while alpha-hydroxy farnesyl phosphonic acid (αHFP) was purchased from Cayman Chemical (Cayman Chemical, MI, USA). Colistin was dissolved in water and αHFP was dissolved in ethanol. All other substances were dissolved in DMSO.

Bacterial strains

All bacterial strains were purchased from Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures. Table 1 lists all used strains alphabetically, giving further information such as the possible affiliation to the ESKAPE group.

Table 1. Alphabetical summary of bacterial strains used in this study.

Bacterial strain Abbreviation ATCC DSM ESKAPEa
Acinetobacter baumannii A. baumannii 19606 30007
Bacillus subtilis strain 168 B. subtilis 23778
Enterobacter cloacae subsp. cloacae E. cloacae 13047 30054
Enterococcus faecium E. faecium 19434 20477
Escherichia coli BW25133 E. coli 27469
Klebsiella pneumoniae subsp. pneumoniae K. pneumoniae 13883 30104
Pseudomonas paraeruginosa b P. paraeruginosa 9027 1128
Staphylococcus aureus S. aureus 25923 1104
Methicillin-resistant Staphylococcus aureus MRSA 33592 11729
Staphylococcus epidermidis S. epidermidis 12228 1798

aESKAPE organisms are indicated with a dot (•).

bThe Pseudomonas strain ATCC 9027 (DSM 1128), previously classified as Pseudomonas aeruginosa, has been reclassified as Pseudomonas paraeruginosa based on recent phylogenetic analyses [20,21].

Antimicrobial susceptibility testing

Broth microdilution assay was used to determine minimal inhibitory concentrations (MICs) of each substance, following CLSI (Clinical and Laboratory Standards Institute) guidelines [22]. For evaluation, resazurin (Sigma Aldrich) was used to indicate bacterial growth [23]. Resazurin is reduced to fluorescent resorufin by viable cells, likely via NADPH dehydrogenase [24]. An Infinite M Plex microplate reader (Tecan Trading AG, Switzerland) measured the fluorescence intensity (excitation at 540 nm, emission at 590 nm). Effective inhibition is assumed if the relative fluorescence intensity (RFI) in all samples is reduced by more than 90% compared to the growth control at a certain concentration, in addition to the visible absence of growth. Minimal bactericidal concentrations (MBCs) were determined immediately after MIC values by plating 3 µl on agar plates without antibacterial agent. Mueller Hinton medium (Carl Roth, Karlsruhe, Germany) was used throughout the experiments, as broth (MHB) or agar (MHA) with 18 g/l agar. The medium was cation-adjusted [25].

Checkerboard assay

In order to evaluate the synergistic effects of the individual substances with colistin, a modified checkerboard assay was used, as described by Bellio, Fagnani [26]. Briefly, a 96-well microplate is divided into two equal parts. This allows two checkerboard assays to be tested on one plate. The dilution scheme and the procedure in general are shown in Fig 2A. Duplicate dilution series for colistin (ranging from 0.008 µg/ml to 0.5 µg/ml for all bacteria except E. cloacae, where concentrations ranged from 0.06 µg/ml to 4.0 µg/ml) and one of each of the other drugs tested (ranging from 7.8 µM to 125.0 µM) were mixed in cation-adjusted MHB. Each well contained 0.2 ml of each antimicrobial combination or broth control. The final bacterial concentration after inoculation was 5 × 105 colony forming units/ml (CFU/ml). Resazurin was used for the indication of bacterial growth, as described in the preceding section (Fig 2B).

Fig 2. Overview of the checkerboard assay.

Fig 2

(A) illustrates the setup process: (1) The appropriate medium, typically MHB, is added to all wells of a 96-well plate. (2) Colistin is added and mixed to the first row, ensuring that the duplicate preparation uses either four or eight times the target concentration, indicated by red or yellow dots. (3) A serial dilution is performed from row A to G, leaving row H free of colistin. (4) The test substance, such as an FTI, is added and mixed to columns 1 and 7, using four times the target concentration. (5) Serial dilutions are then conducted from columns 1 to 5 and 7 to 11, leaving columns 6 and 12 free of the test substance. (6) The plate is inoculated following CLSI guidelines. The color gradients represent the varying concentrations of the substances, while arrows indicate the direction of serial dilutions. The use of a single plate for two tests promotes environmental sustainability. (B) depicts the steps post-incubation: (7) Preliminary results can be observed based on visible bacterial growth or absence thereof. (8) For enhanced visualization, resazurin is added, which is reduced by viable cells to fluorescent resorufin, highlighting cell viability. (9) After a short incubation (1–2 h), fluorescence can be measured for more precise analysis. Additional quantitative assessments may be performed using a microplate reader. Created in BioRender.

The potential synergistic effect of two drugs was calculated using the fractional inhibitory concentration index (FICI) according to the following formula [10]:

FICI= MICA in combinationMICA+ MICB in combinationMICB

In most studies where the checkerboard assay is used, the FICI cutoff to define synergy was set to < 0.5. A range between 0.5 and 1.0 was defined as additive, while 1.0 to 4.0 was interpreted as indifferent. Values of > 4.0 were considered antagonistic [27].

Growth kinetics

To determine the kinetics of bacterial growth, the optical density at 600 nm (OD600) was measured continuously for 24 h at 37°C with shaking at 600 rpm (SPECTROstarNano, BMG Labtech). For visualization, blank-corrected data derived from raw measurements were used. Growth kinetics contribute to understanding the functions of microbial cells and their behavior under different conditions, such as in response to antibiotics [2830].

Time-kill assay

Time-kill assays were conducted to evaluate the bactericidal kinetics and potential synergistic interactions of FTIs and colistin [31]. Bacterial suspensions were adjusted to 0.5 McFarland and diluted 1:200 in MHB to obtain an initial inoculum of 5 × 105 CFU/ml. Cultures were incubated at 37°C with shaking at 180 rpm. At defined time points (0, 1, 2, 4, 8, and 24 h), samples were withdrawn, serially diluted in PBS, and 10 µl were spot plated onto MHA. CFU/ml were determined after overnight incubation at 37°C. Bactericidal activity was defined as a ≥ 3 log10 CFU/ml reduction from the initial inoculum. Synergy was defined as a ≥ 2 log10 CFU/ml reduction compared to the most active single agent [32].

NPN uptake assay

To assess compound-induced changes in outer membrane permeability, N-Phenyl-1-naphthylamine (NPN; Sigma-Aldrich) uptake assays were performed [33,34]. Briefly, bacteria were cultured to exponential phase (OD600 0.3–0.6), harvested, washed twice, and resuspended in assay buffer (5 mM HEPES, 5 mM glucose, pH 7.2) to OD600 1.0. 100 µl of cell suspension were mixed with 100 µl of assay buffer containing 20 µM NPN in black 96-well plates. Fluorescence (excitation 350 nm, emission 420 nm) was monitored every 30 s over 10 min at room temperature. NPN uptake was calculated using the following formula (Muheim et al., 2017), where Fobs is the observed fluorescence at a given compound concentration, Fcon is the fluorescence of NPN with cells plus solvent control, and FB is the fluorescence of NPN in the absence of cells. Data over 10 minutes was averaged.

NPN uptake=(FobsFB)(FconFB)

Statistics

Data processing and statistical analysis were performed using Python 3.12.7 and SciPy 1.14.1 [35]. All data were obtained from at least three independent experiments and presented as means ± standard deviation (SD), unless otherwise noted. For growth kinetics, cubic spline interpolation was applied for data smoothing. For time-kill evaluation, statistical significance was assessed using one-way ANOVA with Tukey’s post hoc test for multiple comparisons. For NPN uptake analysis, one-way ANOVA with Dunnett’s post hoc test was used to compare colistin-treated samples to the control. Significance is indicated as follows: p < 0.05 (*), p < 0.01 (**), p < 0.001 (***).

Results

Antimicrobial susceptibility

To evaluate the FICI and classify the synergistic potential of drugs in combination with colistin, colistin was tested on all strains to determine its MIC (Table 2). In addition, we indicate clinical breakpoints and the respective epidemiological cut-off values (ECOFF), as far as they are provided by the European Committee on Antimicrobial Susceptibility Testing (EUCAST) [36]. Regular antibiotics were tested simultaneously as controls for antimicrobial susceptibility using resazurin as an indicator of cell viability (Fig 3A). Furthermore, gentamicin and vancomycin were tested on S. aureus and methicillin-resistant S. aureus (Fig 3B). RFI values below 10% correspond in all cases with the absence of bacterial growth. All controls are listed in the supplementary files where additional technical information is provided (S1 File).

Table 2. MIC values for colistin determined for the tested bacterial strains.

Bacterial strain Colistin
MIC [µg/ml] Clinical breakpoints ECOFF [µg/ml]
gram-positive B. subtilis 16.0 N/Ab N/A
E. faecium No inhibitiona N/A N/A
S. aureus No inhibition N/A N/A
S. aureus (MRSA) No inhibition N/A N/A
S. epidermidis No inhibition N/A N/A
gram-negative A. baumannii 1.0 Susceptible (≤ 2.0) 2.0
E. cloacae 32.0c Resistant (> 2.0) 2.0
E. coli 0.5 Susceptible (≤ 2.0) 2.0
K. pneumoniae 1.0 Susceptible (≤ 2.0) 2.0
P. paraeruginosa 1.0 Susceptible (≤ 4.0) 4.0d

N/A indicates data not available.

aNo inhibitory effects were observed at the highest tested colistin concentration for E. faecium, S. aureus, methicillin-resistant S. aureus (MRSA), and S. epidermidis.

bEUCAST does not specify clinical breakpoints or ECOFF values for colistin on gram-positive bacteria.

cE. cloacae exhibited a pronounced tendency toward colistin heteroresistance, complicating the determination of a specific MIC.

dFor P. paraeruginosa, the ECOFF value for P. aeruginosa was applied.

Fig 3. Relative fluorescence intensity (RFI) values for the tested bacterial strains and antibiotics.

Fig 3

(A) presents the RFI values for gram-positive and gram-negative bacterial strains treated with varying concentrations of ampicillin, chloramphenicol, kanamycin, and tetracycline. (B) focuses on the RFI values for S. aureus and MRSA treated with gentamicin and vancomycin. A high RFI value indicates active bacterial metabolism and growth, as resazurin is reduced to fluorescent resorufin by viable cells. Conservatively, an RFI below 10% is consistently correlated with the absence of bacterial growth, enabling the assessment of antimicrobial effects and determination of MIC values.

The tested bacterial strains show very diverse sensitivity profiles. In the case of gram-positive bacteria, the difference between S. aureus and MRSA is particularly pronounced. MRSA is resistant to the quantities of ampicillin, chloramphenicol, kanamycin and tetracycline selected in this study. The MRSA strain tested is also less sensitive to gentamicin, while there are no noticeable differences between S. aureus and MRSA with vancomycin. Among the gram-negative strains tested, A. baumannii and P. paraeruginosa are almost exclusively susceptible to colistin and not to the four control antibiotics.

Synergy assessment

When farnesyltransferase inhibitors were combined with sub-inhibitory concentrations of colistin, ranging from 0.008 to 0.5 µg/ml for all bacteria except E. cloacae, where concentrations ranged from 0.06 to 4.0 µg/ml, the growth of gram-positive and gram-negative bacteria was inhibited. The effects of FTIs and colistin were very different in the ten bacterial strains tested. The complete data can be found in the supplementary files (S1S10 Figs).

Among the gram-negative strains, the non-peptidomimetic FTIs lonafarnib and tipifarnib notably reduced the MIC of colistin. For instance, in A. baumannii, the colistin MIC decreased from 1.0 µg/ml to 0.25 µg/ml (four-fold reduction) when 15.6 µM lonafarnib was applied. This effect was even more pronounced with tipifarnib; at 31.3 µM, the colistin MIC was reduced to 0.125 µg/ml (eight-fold reduction). These effects were comparable for E. coli and K. pneumoniae, though the K. pneumoniae strain exhibited a tendency to form subcolonies, particularly noticeable with lonafarnib treatment. Despite this, strong synergy (FICI < 0.5) was observed in some cases. In contrast, P. paraeruginosa largely lacked synergistic interactions, with only tipifarnib showing a slight additive effect. Lonafarnib, even at high concentrations combined with half the colistin MIC, exhibited minimal inhibitory activity against this strain. Conversely, in E. cloacae, lonafarnib displayed stronger synergistic effects than tipifarnib, reducing the colistin MIC from 32 µg/ml to 1.0 µg/ml (32-fold reduction) when 15.6 µM lonafarnib was used.

In comparison, the peptidomimetic FTIs, B581 and FTI-277, exhibited weaker effects on gram-negative bacteria. Additive and partial synergistic effects were observed but were less pronounced than those seen with lonafarnib and tipifarnib. For E. coli, FTI-277 reduced the colistin MIC to 0.125 µg/ml when 31.3 µM was applied. However, in K. pneumoniae, subcolonies reappeared, particularly with FTI-277, leading to inconsistent growth inhibition across replicates and complicating the evaluation of synergy. Similarly, both B581 and FTI-277 showed negligible effects on P. paraeruginosa and E. cloacae.

Finally, αHFP and bempedoic acid exhibited no activity against the gram-negative bacteria tested. Fig 4 summarizes the results for the gram-negative strains, illustrating the investigated properties in relation to the inhibitory effects and identified synergistic activities.

Fig 4. Relative fluorescence intensity (RFI) values for the tested gram-negative bacterial strains and substances, along with corresponding fractional inhibitory concentration index (FICI) values.

Fig 4

The bacterial strains are arranged alphabetically from left to right, with E. cloacae positioned separately on the far right due to the use of different colistin concentrations for this strain. FICI values are visualized to classify interactions: values below 0.5 indicate strong synergy, values between 0.5 and 1.0 represent additive effects, and values from 1.0 to 4.0 suggest indifference. No antagonistic interactions (FICI > 4.0) were observed. The RFI values for αHFP and bempedoic acid (BPD) are only partially displayed, as these substances demonstrated no inhibitory effects.

The tested gram-positive bacteria differ significantly from the gram-negative strains. No effects of B581, FTI-277, αHFP, or BPD were observed in any of the gram-positive representatives. However, lonafarnib and tipifarnib exhibited strong inhibitory effects on MRSA, S. aureus, and S. epidermidis, independent of colistin. Thus, no synergistic effects were identified in these cases and colistin did not compromise the inhibitory effects of lonafarnib or tipifarnib on these bacterial strains.

MRSA was inhibited at a lonafarnib concentration of 31.3 µM, whereas non-methicillin-resistant S. aureus showed inhibition at 15.6 µM. Similar MIC values were observed for tipifarnib against these strains; however, S. aureus exhibited inconsistent behavior with tipifarnib. At lower concentrations, tipifarnib partially reduced growth without achieving complete inhibition. Both lonafarnib and tipifarnib affected S. epidermidis similarly. Conversely, E. faecium showed no growth inhibition with either lonafarnib or tipifarnib when standard microdilution methods were applied. It is important to note that the E. faecium strain used in this study displayed an overall slow growth rate, which was further reflected in its generally low metabolic activity, as indicated by a slower reduction of resazurin.

A unique case among the gram-positive bacteria was B. subtilis. This was the only gram-positive organism successfully inhibited by colistin (16 µg/mL) in this study. Furthermore, lonafarnib exhibited inhibitory effects on B. subtilis growth only in combination with colistin. A successful inhibition was observed with 31.3 µM lonafarnib and 0.5 µg/mL colistin, representing a 32-fold reduction in the required colistin concentration. Since colistin alone demonstrated activity against B. subtilis, an additive effect with tipifarnib and colistin is inferred. At a tipifarnib concentration of 15.6 µM, the colistin exhibited measurable effects. These findings are summarized in Fig 5.

Fig 5. Relative fluorescence intensity (RFI) values for the tested gram-positive bacterial strains and substances, along with fractional inhibitory concentration index (FICI) values.

Fig 5

The bacterial strains are arranged alphabetically from left to right. FICI values are visualized for B. subtilis, with no antagonistic effects observed (FICI > 4.0). The RFI values for B581, FTI-277, αHFP, and bempedoic acid (BPD) are only partially displayed, as these substances showed no inhibitory effects.

To summarize the findings across all bacterial strains, the overall highest FICI values and fold changes in colistin MIC are presented as heatmaps in Fig 6. These data illustrate strain-specific variations in FTI/colistin interactions and colistin MIC reductions.

Fig 6. Heatmap comparison of the highest fractional inhibitory concentration index (FICI) values and fold change (FC) reductions in colistin MIC across all tested bacterial strains.

Fig 6

(A) depicts strain-specific synergy between FTIs and colistin, with lonafarnib and tipifarnib exhibiting the strongest effects against gram-negative bacteria such as E. cloacae and K. pneumoniae. (B) highlights fold changes in colistin MIC, showcasing substantial reductions, including a 32-fold decrease in B. subtilis (with lonafarnib) and E. cloacae (with both lonafarnib and tipifarnib). These heatmaps provide an overview of differential bacterial responses, emphasizing the potential of FTI/colistin combinations to enhance antimicrobial efficacy and reduce colistin requirements.

Lonafarnib and tipifarnib were the only FTIs that exhibited effects against gram-positive bacteria. However, synergistic effects (FICI < 0.5) were observed exclusively in B. subtilis, while E. faecium remained unaffected by any of the tested substances under the given conditions. In contrast, all tested FTIs inhibited gram-negative bacteria, with lonafarnib and tipifarnib demonstrating the highest FICI values in combination with colistin, particularly against E. cloacae and K. pneumoniae (FICI < 0.25). P. paraeruginosa was the least affected among the gram-negative strains (Fig 6A).

Within the gram-positive bacterial strains tested in this study, only B. subtilis exhibited a change in the required colistin MIC. A 32-fold reduction in the colistin concentration was observed when lonafarnib was administered concurrently. Although a certain additive effect was also observed with the simultaneous administration of colistin and tipifarnib, no fold change (FC) in colistin MIC was reported. This is because tipifarnib, at a concentration of 31.3 µM, inhibited the growth of B. subtilis independently of colistin. For gram-negative bacteria, FICI values strongly correlated with colistin MIC FCs. The most pronounced effect was observed in E. cloacae, where lonafarnib and tipifarnib both reduced the required colistin concentration from 32.0 µg/ml to 1.0 µg/ml, corresponding to a 32-fold reduction in colistin MIC in a concentration-dependent manner. In contrast, peptidomimetic FTIs failed to reduce the colistin MIC in E. cloacae. This was not the case for other gram-negative strains, where reductions were observed. Yet, K. pneumoniae was affected by B581 but not by FTI-277 (Fig 6B).

Growth kinetics and time-kill

While the checkerboard assays explored the synergistic effects of FTIs and colistin across both gram-positive and gram-negative bacteria, growth kinetics experiments were conducted to gain deeper insights into the temporal dynamics of FTI activity, focusing specifically on S. aureus, a clinically significant gram-positive pathogen, and E cloacae, a colistin resistant strain. These experiments aimed to complement the endpoint-based MIC and FICI results by evaluating the consistency and duration of growth inhibition under dynamic conditions. To this end, S. aureus was tested with four concentrations of lonafarnib and tipifarnib, respectively, using continuous OD600 measurements over 24 h at 37°C with shaking at 600 rpm. These tests revealed slight deviations compared to standard CLSI-based methods. At a concentration of 7.8 µM lonafarnib, the growth of S. aureus was completely inhibited for the first 16 h, after which rapid bacterial growth resumed (Fig 7A). Higher concentrations of 15.6 µM and 31.3 µM lonafarnib resulted in continuous growth inhibition throughout the 24-hour measurement period. A similar pattern was observed for tipifarnib: at 7.8 µM, minimal growth was detectable as a slight increase in OD600 after 12 h (Fig 7C). Higher concentrations also led to sustained inhibition of growth. The observed differences may indicate an influence of shaking on the experimental outcomes, particularly regarding compound distribution in the medium.

Fig 7. Growth kinetics and time-kill assays of methicillin-sensitive Staphylococcus aureus and colistin-resistant Enterobacter cloacae.

Fig 7

Growth kinetics are shown as the mean optical density (OD) at 600 nm plotted against different substance concentrations or combinations, with each concentration of combination represented by a different color. Standard deviation (SD) values are indicated as shaded areas around the primary plot lines. Cubic spline interpolation was applied to smooth the data. Panels (A) and (C) show the growth kinetics for S. aureus, while panels (E) and (G) display the results for E. cloacae. To assess bactericidal activity over time, time-kill assays were performed. Panels (B) and (D) show the time-kill results for S. aureus, and panels (F) and (H) for E. cloacae. Statistical analysis was conducted using one-way ANOVA with Tukey’s post hoc test for multiple comparisons. Significance is indicated as follows: p < 0.05 (*), p < 0.01 (**), p < 0.001 (***). A dashed line at log10 2 indicates the limit of detection, corresponding to 100 CFU/ml. Values below this threshold could not be reliably quantified due to technical reasons. For the time-kill assays, lonafarnib and tipifarnib were assessed simultaneously. Visualization was split to allow better distinction. This resulted in shared controls for panels (B) and (D), and for panels (F) and (H).

Growth kinetics were similarly assessed for E cloacae. Unlike S. aureus, growth inhibition was not observed with lonafarnib or tipifarnib alone at concentrations up to 31.3 µM, or with sub-inhibitory concentrations of colistin. However, clear growth suppression was observed in combination treatments with 4 µg/ml of colistin and 15.6 µM or 31.3 µM of lonafarnib, which maintained growth inhibition for the entire 24-hour measurement period. Even a concentration of 7.8 µM of lonafarnib combined with colistin resulted in a transient growth delay compared to colistin alone (Fig 7E). Tipifarnib/sub-inhibitory colistin had a similar effect on growth kinetics, albeit less pronounced. Growth was prevented over 24 h at 31.3 µM tipifarnib plus 4 µg/ml colistin, while combinations at 15.6 µM caused a delayed onset of growth by about 10 h (Fig 7G).

Time-kill assays were conducted to gain more insights into the bactericidal kinetics of FTIs and colistin. Due to S. aureus’s inherently slower growth rate compared to E. cloacae, measurable differences in bacterial counts emerged after 4 h. By 8 h, all tested concentrations of lonafarnib had resulted in a significant reduction of over 3 log10 CFU/ml compared to the DMSO control (p < 0.001) (Fig 7B). After 24 h, this reduction remained significant across all concentrations, although the effect of 7.8 µM was notably less pronounced. A similar effect was observed for tipifarnib: while all tested concentrations demonstrated bactericidal activity, the highest concentration (31.3 µM) led to a reduction of approximately 7 log10 CFU/ml compared to the control (p < 0.0001). There was a statistically significant difference between the 31.3 µM and 15.6 µM concentrations of tipifarnib (p < 0.0001), indicating a concentration-dependent effect (Fig 7D).

For E. cloacae, the results for lonafarnib (Fig 7F) and tipifarnib (Fig. 7H) were highly comparable. Neither compound exhibited any bactericidal activity when used alone, with no reduction in CFU/ml relative to the DMSO control. Colistin, when applied at sub-inhibitory concentrations, induced a transient reduction in bacterial counts, most notably at the earliest time points. After 4 h, treatment with colistin alone reduced bacterial load by approximately 5 log10 CFU/ml compared to the control (p < 0.001). However, this effect was not sustained; by 8 h, the reduction no longer reached the ≥ 2 log10 CFU/ml threshold for bactericidal activity, and by 24 h, CFU levels had returned to control levels. In contrast, the combined approach demonstrated clear synergy. When combined with sub-inhibitory concentrations of colistin, both lonafarnib and tipifarnib resulted in a ≥ 3 log10 CFU/ml reduction compared to colistin alone, fulfilling the criterion for synergistic bactericidal activity. These differences were highly significant at the 2-hour time point for both combinations (p < 0.0001).

Outer membrane permeability

Focusing on the outer membrane of gram-negative bacteria, permeability was assessed for E. cloacae and E. coli using the fluorescent dye NPN. This small molecule exhibits low fluorescence in an aqueous solution but becomes strongly fluorescent when it binds to phospholipids. Under normal conditions, uptake is limited and fluorescence remains low. However, when the OM is compromised, NPN can penetrate more easily. This results in increased fluorescence due to its interaction with membrane lipids [33]. The data were normalized to the highest tested colistin concentration (2 × MIC).

In E. cloacae, all tested colistin concentrations – including sub-inhibitory levels down to 1/8 MIC – led to a highly significant increase in NPN uptake compared to the DMSO control (p < 0.0001 for all conditions). While the increased fluorescence at 1/2 MIC and 1/4 MIC could be partly explained by cell lysis, this is unlikely at 1/8 MIC, despite the NPN signal remaining significantly elevated (Figs 8AB). For E. coli, all tested concentrations – including sub-inhibitory levels – resulted in a statistically significant increase in NPN uptake compared to the DMSO control (p < 0.0001 to p = 0.0087) (Figs 8CD). These findings indicate that even sub-inhibitory concentrations of colistin can increase outer membrane permeability in both species.

Fig 8. Sub-inhibitory concentrations of colistin increase outer membrane permeability in E. cloacae (A) and E. coli (C).

Fig 8

The integrity of the outer membrane (OM) was assessed using the hydrophobic fluorescent probe N-phenyl-1-naphthylamine (NPN), which does not invade cells with an intact OM, but fluoresces when inserted into the phospholipid bilayer upon OM compromise. The effect of different concentrations of colistin on OM permeability was determined. Data were normalized to the 2 × MIC colistin condition. DMSO served as a negative control and H2O as the colistin solvent. Statistical analysis was conducted using one-way ANOVA with Dunnett’s post hoc test for multiple comparisons. Significance is indicated as follows: p < 0.05 (*), p < 0.01 (**), p < 0.001 (***). The sub-inhibitory concentration range is indicated by a light grey box (“sub-MIC”). To control for antimicrobial activity, dilution series were spotted on MHA to confirm that sub-inhibitory concentrations of colistin alone do not inhibit the growth of E. cloacae (B) and E. coli (D).

Discussion

In this study, the effects of different farnesyltransferase inhibitors in combination with colistin were investigated to identify potential synergistic effects and expand the spectrum of FTIs to gram-negative bacteria. The main purpose of FTIs is to inhibit FTase, an enzyme that, in eukaryotic cells, catalyzes the post-translational transfer of a C15 (farnesyl) isoprenoid moiety from farnesyl pyrophosphate (FPP) to a cysteine residue in target protein [37]. Together with geranylgeranyltransferases, FTase plays a central role in protein prenylation [38,39].

The formation of isoprenoids, necessary for the prenylation, such as FPP and geranylgeranyl pyrophosphate (GGPP), relies on either the mevalonate pathway or, more commonly in bacteria, the methylerythritol phosphate (MEP) pathway [40,41]. Independently of the biosynthetic pathway used, isoprenoids are universally distributed across all domains of life – bacteria, archaea, and eukaryotes – and are essential for cell survival, including in bacterial pathogens [42]. This universality underscores their critical role in various cellular processes. Interestingly, the antimicrobial effects of FTIs appear to be independent of the biosynthetic pathway for isoprenoid production, as FTIs effectively inhibit the growth of bacteria utilizing both the MEP and mevalonate pathways [43]. Isoprenoids are vital for bacterial cells, serving roles in pathogenicity [44,45] and acting as intermediates in cell wall biosynthesis [46]. However, their utilization in protein prenylation within bacterial cells remains largely elusive [5].

As a ubiquitous covalent post-translational modification in eukaryotic cells, protein prenylation involves the attachment of isoprenoids like FPP or GGPP to proteins [47]. In contrast, bacteria rarely employ such modifications, as post-translational modifications are generally less prevalent in prokaryotes [48]. The extent of bacterial protein modification varies significantly among species and is strongly influenced by environmental conditions. Nevertheless, post-translational modifications can profoundly impact bacterial physiology and virulence regulation, altering protein structure, activity, localization, and biomolecular interactions [49]. While no evidence suggests that bacteria utilize an inherent protein prenylation machinery for post-translational modifications, some pathogenic bacteria exploit host-cell prenylation systems. For instance, bacterial effector proteins with a C-terminal CAAX motif have been shown to be prenylated by the host’s prenylation machinery. This phenomenon has been observed in pathogens such as Salmonella typhimurium and Legionella pneumophila [5052]. Additionally, several other bacterial proteins, primarily from pathogenic strains, have been identified as potential targets for host-mediated prenylation, including possible farnesylation [53]. While endogenous bacterial protein prenylation remains largely unexplored, some exceptions exist. For instance, B. subtilis utilizes a distinct prenylation system for the quorum-sensing protein ComX, where a farnesyl group is transferred to a tryptophan residue by ComQ, rather than by FTase [54]. Notably, the antimicrobial effects observed in our study are also independent of host prenylation systems, suggesting that bacterial prenylation-related mechanisms may be relevant targets. Nonetheless, the reliance of these pathogens on host prenyltransferases does not preclude the possibility that bacterial prenyltransferases may also exist [5]. Therefore, the development of FTIs, which is already conducted in the field of cancer therapy, could also be of great importance in the treatment of infectious diseases of various kinds. For instance, lonafarnib is under clinical investigation for the treatment of hepatitis D virus (HDV) infections. Additionally, lonafarnib shows promising effects for managing bronchiolitis and pneumonia caused by respiratory syncytial virus (RSV) and has also demonstrated effects against SARS-CoV-2 [5557]. Recent studies have also highlighted the antimicrobial effects of FTIs. We demonstrated significant antimicrobial activity of lonafarnib and tipifarnib – two non-peptidomimetic FTIs – against gram-positive bacteria, including S. aureus, S. epidermidis, and even methicillin-resistant S. aureus. However, no activity of these compounds against gram-negative bacteria was reported, such as E. coli, K. pneumoniae, and P. aeruginosa. It was hypothesized that the lack of activity was due to the OM of gram-negative bacteria, which serves as an additional intrinsic permeability barrier that is absent in gram-positive bacteria [5].

In the present study, colistin, a well-established antimicrobial from the polymyxin class, was used to overcome this limitation in order to use the FTIs to treat a broad range of bacterial strains. Colistin disrupts the OM of gram-negative bacteria by binding to lipopolysaccharides (LPS) via electrostatic interactions, ultimately leading to bacterial cell death [8,58]. When sub-inhibitory concentrations of colistin were applied to compromise the OM, lonafarnib and tipifarnib exhibited significant antimicrobial effects against gram-negative bacteria. Additionally, this approach enabled the peptidomimetic FTIs, B581 and FTI-277, to inhibit the growth of gram-negative bacterial strains. Interestingly, these two peptidomimetic FTIs did not show any activity against gram-positive bacteria, in contrast to lonafarnib and tipifarnib. This difference is likely attributable to their distinct chemical properties, despite sharing the same enzymatic target in eukaryotic cells. Notably, lonafarnib and tipifarnib are halogenated, whereas the peptidomimetic FTIs are not. Halogenated compounds are known to exhibit increased affinity for gram-positive bacteria [59], which may explain the observed activity patterns, and should be further studied. The strategy of using sub-inhibitory concentrations of colistin is well-established in the field of drug repurposing to evaluate the antimicrobial activity of compounds against gram-negative bacteria. Statins, for example, which are widely used as lipid-modifying agents to control hyperlipidemia, have demonstrated antimicrobial properties [4]. This unexpected effect was initially observed as a pleiotropic property of these drugs. Epidemiological studies further suggested a correlation between statin therapy and a reduced risk of sepsis and other severe bacterial infections [60,61]. Initial susceptibility testing revealed that statins exhibit antimicrobial activity, though the clinical relevance remains uncertain. This is because the in vivo concentrations achieved with clinical statin doses are significantly lower than those required for antimicrobial activity [62]. Nonetheless, subsequent studies have highlighted the potential of statins as novel antimicrobials and demonstrated their synergistic effects even on gram-negative bacteria when combined with colistin [63,64]. Although the mechanism of action (MOA) of statins in bacteria is still not fully elucidated, in humans, statins exert their primary effect by inhibiting class I HMG-CoA reductase within the mevalonate pathway [65]. However, only a few bacterial species utilize the mevalonate pathway to produce isopentenyl pyrophosphate and dimethylallyl pyrophosphate [66,67]. Consequently, Thangamani, Mohammad [64] hypothesized that the MOA of simvastatin, a representative statin with notable antimicrobial activity, differs in bacteria due to the absence of the class I HMG-CoA reductase enzyme. Instead, its MOA appears to be complex, involving inhibition of multiple biosynthetic pathways and cellular processes, including selective interference with bacterial protein synthesis [64]. This complexity may be partially adaptable to FTIs, as statins target an earlier step in the mevalonate pathway, upstream of the isoprenoid synthesis required for processes like prenylation. Even further upstream of the mevalonate pathway, another inhibitor that was used in this study, bempedoic acid, exerts its effect. However, bempedoic acid showed no measurable effect on the growth of the bacterial strains tested in this study, regardless of the presence of sub-inhibitory colistin. Bempedoic acid is known to lower low-density lipoprotein cholesterol levels in humans by inhibiting ATP citrate lyase, a key enzyme in the cholesterol biosynthesis pathway that acts upstream of HMG-CoA reductase, the target of statins [19]. While ATP citrate lyase plays a crucial role in autotrophic organisms, such as green sulfur bacteria, by facilitating carbon dioxide fixation [68,69], the gram-positive and gram-negative bacteria tested in this study do not rely on this enzyme for survival.

Furthermore, another inhibitor tested in this study, αHFP, a nonhydrolyzable analog of FPP, functions as a competitive inhibitor of FTase in eukaryotic cells and effectively blocks Ras farnesylation [70,71]. This compound was included in the study to provide a comparative context for the activity of FTIs, as αHFP mimics FPP, the natural substrate of FTase. While αHFP has demonstrated significant effects in eukaryotic systems [72], none of the bacterial strains in this study responded to its presence. This observation highlights the distinct mechanisms of action of FTIs in eukaryotic versus bacterial cells and emphasizes the need for further investigation into their activity in different biological systems.

Further investigation into the effects of FTIs on B. subtilis, an important model organism, could be pivotal in elucidating their MOA, particularly regarding interactions with the bacterial cell membrane and wall. This study found that B. subtilis strain 168 is not only susceptible to colistin at relatively high concentrations but is also the only gram-positive bacterium among those tested to exhibit a unique inhibition pattern for drug combinations. Colistin likely acts on B. subtilis by stimulating both the TCA cycle and the respiratory chain, enhancing NADH metabolism and leading to oxidative damage, a mechanism that could potentially be applicable to other gram-positive bacteria [73]. While lonafarnib demonstrated a strong synergistic effect only in combination with colistin, tipifarnib appeared to act mostly independently. Notably, this specific strain has been extensively used in research for many years [74]. Recently, Leeten, Jacques [75] highlighted the pleiotropic antimicrobial effects of the antiplatelet drug ticagrelor, employing B. subtilis strain 168 in assays specifically designed to study cell wall and membrane alterations.

Although bacteria seem to lack their own prenylation machinery, these findings suggest that host-pathogen interactions involving protein prenylation play a significant role in bacterial virulence. This highlights the broader importance of understanding prenylation in bacterial biology and its potential as a therapeutic target [76]. Building on this concept, the tested FTIs – non-peptidomimetic lonafarnib and tipifarnib, as well as peptidomimetic B581 and FTI-277 – demonstrated the ability to inhibit bacterial growth in this study. While the exact MOA of FTIs in bacteria remains unclear, the observed activity supports the hypothesis that their effects differ from their MOA in eukaryotic cells, potentially involving a complex mechanism with multiple targets that disrupt various biosynthetic pathways and cellular processes. Importantly, these findings demonstrate that FTIs, when combined with sub-inhibitory concentrations of colistin, can extend their antimicrobial activity to gram-negative bacteria. Fig 9 illustrates the current understanding of this process: colistin disrupts the OM of gram-negative bacteria, enabling FTIs to penetrate and exert a concentration-dependent effect on bacterial cells.

Fig 9. Proposed mechanism of action for farnesyltransferase inhibitors (FTIs) in combination with colistin on bacteria.

Fig 9

At sub-inhibitory concentrations, colistin disrupts the outer membrane (OM) of gram-negative bacteria by binding to Lipid A in the lipopolysaccharide, compromising the integrity of the OM. This disruption allows FTIs to access their target(s), which remain unidentified. Potential mechanisms include interference with multiple biosynthetic pathways, or disruption of prenylation processes within the bacterial cell. These combined effects ultimately result in bacterial cell death. Created in BioRender.

FTIs not only demonstrate the ability to inhibit gram-positive and gram-negative bacteria but also significantly reduce the MIC of colistin in these strains. This includes the E. cloacae strain used in this study, a member of the C-XI cluster with intrinsic colistin resistance and an associated heteroresistance phenotype [77,78]. MIC values for colistin in this strain vary widely, with this study estimating a value that falls within the expected range for colistin-resistant strains (Table 1). It is important to note that, even under optimal conditions, MIC values should be interpreted as approximate ranges rather than precise figures, as variability in measurement is inherent to the method (EUCAST v. 14). However, the strong synergistic effect observed with the combined use of colistin and FTIs supports the hypothesis that FTIs may provide insights into overcoming the growing resistance to colistin. As a member of the ESKAPE pathogen group, E. cloacae is responsible for numerous opportunistic nosocomial infections and, less frequently, community-acquired infections. These include urinary tract infections, bacteremia, lower respiratory tract infections, and surgical site infections, as well as colonization of intravascular devices [79]. The E. cloacae strain used in this study, which is also the type strain of E. cloacae subsp. cloacae, exhibits high levels of colistin resistance and is resistant to ampicillin (Fig 3). This strain also demonstrates significant pathogenic potential due to several mechanisms and can, for example, harbor elements such as a type VI secretion system, which is crucial for survival within the host [80,81]. In this study, lonafarnib and tipifarnib reduced the colistin MIC in E. cloacae by more than 32-fold (Fig 6). Furthermore, the highest FICI values for synergy were calculated for E. cloacae and K. pneumoniae, using a conservative approach. Specifically, the MIC values of FTIs were set to the highest tested concentrations, as no inhibitory effect was observed without the simultaneous use of colistin in gram-negative strains. This conservative methodology likely underestimates the true extent of synergy, as higher individual MIC values for FTIs would yield even more pronounced synergistic FICI values. However, the approach used in this study aligns with previous studies that assessed FICI values for substances exhibiting no standalone effect on gram-negative bacteria, such as baicalin, flavomycin, and resveratrol [10,82].

Beyond the already demonstrated potential of FTIs as antimicrobial agents against gram-positive and gram-negative bacteria, this study also highlights their ability to reduce the MIC of colistin for colistin-resistant bacteria. As the prevalence of colistin-resistant strains continues to rise, colistin – once discontinued due to its neurotoxicity and nephrotoxicity – has been reinstated as a “last-resort” antibiotic for treating infections caused by gram-negative nosocomial pathogens such as A. baumannii [83]. The findings of this study may have broader implications for addressing colistin-resistant bacteria. To evaluate this hypothesis, further testing on diverse colistin-resistant strains, including those harboring mobile colistin resistance genes, is warranted. Additionally, this approach could target bacteria with a strong propensity for heteroresistance to colistin and other polymyxins. Although heteroresistance can occur in both gram-positive and gram-negative bacteria, its increasing prevalence in gram-negative pathogens such as E. coli and K. pneumoniae is of particular concern [84]. In this study, the tendency of K. pneumoniae to form subcolonies appeared to be diminished when tipifarnib was combined with colistin, suggesting that tipifarnib may contribute to reducing the MIC of colistin and potentially affect heteroresistance in this strain. Further research should be conducted to determine whether the observed inhibition patterns are attributable to the modulation of heteroresistance or rather reflect compound-specific characteristics [85].

For any potential therapeutic use of FTIs as antimicrobial agents in humans, whether as adjuvants or monotherapies, in vivo concentrations would need to approach MIC values. For gram-negative strains, the highest FICI values were observed at FTI concentrations of 15.6 µM to 31.3 µM, although even lower concentrations, such as 7.8 µM, demonstrated a reduction in colistin MIC. For example, in a study involving patients with advanced solid tumors, lonafarnib achieved a maximum plasma concentration of 3.8 µg/ml after administering 300 mg twice daily [86], equivalent to approximately 6.0 µM. While such high doses of an anticancer drug are unlikely to be used for treating infections, this scenario might be feasible in cases of severe healthcare-associated infections. Beyond our in vitro findings, animal models could provide a more realistic representation of both the pharmacokinetics and pharmacodynamics of FTIs, offering valuable insights into their in vivo efficacy and antimicrobial potential. Future studies should incorporate diverse testing media and preclinical models to validate and build upon the results observed in vitro. These approaches could also elucidate the impact of FTIs on the human gut microbiome – an important consideration for systemic use – given its potential relevance in cancer therapy [87]. It should also be considered that bacteria may metabolize FTIs, although the specific metabolites remain unknown. Furthermore, even if systemic plasma concentrations remain insufficient, FTIs could have potential as topical agents, allowing for higher localized concentrations in specific areas, as has been shown for statins before [64].

Conclusion

In summary, this basic research explored the effects of FTIs and additional compounds on key pathogenic bacterial strains in combination with sub-inhibitory concentrations of colistin. The non-peptidomimetic FTIs, lonafarnib and tipifarnib, demonstrated significant activity against gram-negative bacteria when co-administered with sub-inhibitory concentrations of colistin, extending their activity beyond gram-positive bacteria. Notably, FTIs were able to reduce colistin MICs, even in a colistin-resistant gram-negative strain. Peptidomimetic FTIs, B581 and FTI-277, were also effective against gram-negative bacteria but exhibited no activity against gram-positive strains. In contrast, αHFP and bempedoic acid showed no effect on any tested strains, supporting the hypothesis that the mechanism of action of FTIs differs substantially from their activity in eukaryotic cells. The observed effects are likely due to the disruption of multiple biosynthetic pathways, underscoring the potential of FTIs as adjunctive drugs in antimicrobial therapy. Further studies will be conducted to clarify the mechanisms and potential of FTIs as pleiotropic antimicrobial substances, potentially making a leap in combating the ever-increasing threat of resistant pathogens.

Supporting information

S1 File. Additional information and controls.

The file provides supporting information on the Tecan device and the program used for fluorescence measurements, as well as details of the conducted controls.

(DOCX)

pone.0331440.s001.docx (1.2MB, docx)
S1 Fig. RFI values for A. baumannii.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for A. baumannii. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s002.tif (468.3KB, tif)
S2 Fig. RFI values for B. subtilis.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for B. subtilis. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s003.tif (464.7KB, tif)
S3 Fig. RFI values for E. cloacae.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for E. cloacae. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s004.tif (451.3KB, tif)
S4 Fig. RFI values for E. faecium.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for E. faecium Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s005.tif (477.9KB, tif)
S5 Fig. RFI values for E. coli.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for E. coli. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s006.tif (462.1KB, tif)
S6 Fig. RFI values for K. pneumoniae.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for K. pneumoniae. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s007.tif (468.4KB, tif)
S7 Fig. RFI values for MRSA.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for MRSA. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s008.tif (459.3KB, tif)
S8 Fig. RFI values for P. paraeruginosa.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for P. paraeruginosa. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s009.tif (467.4KB, tif)
S9 Fig. RFI values for S. aureus.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for S. aureus. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s010.tif (461.6KB, tif)
S10 Fig. RFI values for S. epidermidis.

Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for S. epidermidis. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

(TIF)

pone.0331440.s011.tif (466.8KB, tif)

Acknowledgments

The authors would like to thank Tim Bürgel for his advice and introduction to image creation with BioRender and Raffaela Maltaner for her technical support in the laboratory.

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

The author(s) received no specific funding for this work.

References

  • 1.De Oliveira DMP, et al. Antimicrobial Resistance in ESKAPE Pathogens. Clin Microbiol Rev. 2020;33(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gigante V, Sati H, Beyer P. Recent advances and challenges in antibacterial drug development. ADMET DMPK. 2022;10(2):147–51. doi: 10.5599/admet.1271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Huemer M, Mairpady Shambat S, Brugger SD, Zinkernagel AS. Antibiotic resistance and persistence-Implications for human health and treatment perspectives. EMBO Rep. 2020;21(12):e51034. doi: 10.15252/embr.202051034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hennessy E, Adams C, Reen FJ, O’Gara F. Is There Potential for Repurposing Statins as Novel Antimicrobials? Antimicrob Agents Chemother. 2016;60(9):5111–21. doi: 10.1128/AAC.00192-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Weber L, et al. Bacteria are new targets for inhibitors of human farnesyltransferase. Front Microbiol. 2021;12:628283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Reshamwala SMS, Mamidipally C, Pissurlenkar RRS, Coutinho EC, Noronha SB. Evaluation of risedronate as an antibiofilm agent. J Med Microbiol. 2016;65(1):9–18. doi: 10.1099/jmm.0.000193 [DOI] [PubMed] [Google Scholar]
  • 7.Kruszewska H, Zareba T, Tyski S. Examination of antimicrobial activity of selected non-antibiotic medicinal preparations. Acta Pol Pharm. 2012;69(6):1368–71. [PubMed] [Google Scholar]
  • 8.El-Sayed Ahmed MAE, et al. Colistin and its role in the era of antibiotic resistance: an extended review (2000-2019). Emerg Microbes Infect. 2020;9(1):868–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rhouma M, Beaudry F, Letellier A. Resistance to colistin: what is the fate for this antibiotic in pig production?. Int J Antimicrob Agents. 2016;48(2):119–26. [DOI] [PubMed] [Google Scholar]
  • 10.Huang Y, Zhu Y, Yue H-Y, Liu Y-Y, Deng L-M, Lv L, et al. Flavomycin restores colistin susceptibility in multidrug-resistant Gram-negative bacteria. mSystems. 2024;9(6):e0010924. doi: 10.1128/msystems.00109-24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rice LB. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J Infect Dis. 2008;197(8):1079–81. doi: 10.1086/533452 [DOI] [PubMed] [Google Scholar]
  • 12.Miller WR, Arias CA. ESKAPE pathogens: antimicrobial resistance, epidemiology, clinical impact and therapeutics. Nat Rev Microbiol. 2024;22(10):598–616. [DOI] [PubMed] [Google Scholar]
  • 13.Boucher HW, et al. Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of America. Clin Infect Dis. 2009;48(1):1–12. [DOI] [PubMed] [Google Scholar]
  • 14.Lorusso AB, et al. Role of efflux pumps on antimicrobial resistance in Pseudomonas aeruginosa. Int J Mol Sci. 2022;23(24). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Savin M, et al. Tracing clinically-relevant antimicrobial resistances in Acinetobacter baumannii-calcoaceticus complex across diverse environments: A study spanning clinical, livestock, and wastewater treatment settings. Environ Int. 2024;186:108603. [DOI] [PubMed] [Google Scholar]
  • 16.Jones RM. The influence of the gut microbiota on host physiology: in pursuit of mechanisms. Yale J Biol Med. 2016;89(3):285–97. [PMC free article] [PubMed] [Google Scholar]
  • 17.DeGruttola AK, et al. Current understanding of dysbiosis in disease in human and animal models. Inflamm Bowel Dis. 2016;22(5):1137–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dhillon S. Lonafarnib: First approval. Drugs. 2021;81(2):283–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ray KK, Bays HE, Catapano AL, Lalwani ND, Bloedon LT, Sterling LR, et al. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N Engl J Med. 2019;380(11):1022–32. doi: 10.1056/NEJMoa1803917 [DOI] [PubMed] [Google Scholar]
  • 20.Rudra B, et al. Phylogenomic and comparative genomic studies robustly demarcate two distinct clades of Pseudomonas aeruginosa strains: proposal to transfer the strains from an outlier clade to a novel species Pseudomonas paraeruginosa sp. nov. Int J Syst Evol Microbiol. 2022;72(11). [DOI] [PubMed] [Google Scholar]
  • 21.Rudra B, et al. Corrigendum: Phylogenomic and comparative genomic studies robustly demarcate two distinct clades of Pseudomonas aeruginosa strains: proposal to transfer the strains from an outlier clade to a novel species Pseudomonas paraeruginosa sp. nov. Int J Syst Evol Microbiol. 2023;73(3). [DOI] [PubMed] [Google Scholar]
  • 22.Humphries R, Bobenchik AM, Hindler JA, Schuetz AN. Overview of Changes to the Clinical and Laboratory Standards Institute Performance Standards for Antimicrobial Susceptibility Testing, M100, 31st Edition. J Clin Microbiol. 2021;59(12):e0021321. doi: 10.1128/JCM.00213-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Elshikh M, Ahmed S, Funston S, Dunlop P, McGaw M, Marchant R, et al. Resazurin-based 96-well plate microdilution method for the determination of minimum inhibitory concentration of biosurfactants. Biotechnol Lett. 2016;38(6):1015–9. doi: 10.1007/s10529-016-2079-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Costa P, Gomes ATPC, Braz M, Pereira C, Almeida A. Application of the Resazurin Cell Viability Assay to Monitor Escherichia coli and Salmonella Typhimurium Inactivation Mediated by Phages. Antibiotics. 2021;10(8):974. doi: 10.3390/antibiotics10080974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wiegand I, Hilpert K, Hancock REW. Agar and broth dilution methods to determine the minimal inhibitory concentration (MIC) of antimicrobial substances. Nat Protoc. 2008;3(2):163–75. doi: 10.1038/nprot.2007.521 [DOI] [PubMed] [Google Scholar]
  • 26.Bellio P, Fagnani L, Nazzicone L, Celenza G. New and simplified method for drug combination studies by checkerboard assay. MethodsX. 2021;8:101543. doi: 10.1016/j.mex.2021.101543 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Balouiri M, Sadiki M, Ibnsouda SK. Methods for in vitro evaluating antimicrobial activity: A review. J Pharm Anal. 2016;6(2):71–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Fernandez-Martinez LT, Javelle A, Hoskisson PA. Microbial primer: Bacterial growth kinetics. Microbiology (Reading). 2024;170(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Iyer V, Raut J, Dasgupta A. Impact of pH on growth of Staphylococcus epidermidis and Staphylococcus aureus in vitro. J Med Microbiol. 2021;70(9). [DOI] [PubMed] [Google Scholar]
  • 30.Myers JA, Curtis BS, Curtis WR. Improving accuracy of cell and chromophore concentration measurements using optical density. BMC Biophys. 2013;6(1):4. doi: 10.1186/2046-1682-6-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pankey GA, Sabath LD. Clinical relevance of bacteriostatic versus bactericidal mechanisms of action in the treatment of Gram-positive bacterial infections. Clin Infect Dis. 2004;38(6):864–70. doi: 10.1086/381972 [DOI] [PubMed] [Google Scholar]
  • 32.Belley A, Neesham-Grenon E, Arhin FF, McKay GA, Parr TR Jr, Moeck G. Assessment by time-kill methodology of the synergistic effects of oritavancin in combination with other antimicrobial agents against Staphylococcus aureus. Antimicrob Agents Chemother. 2008;52(10):3820–2. doi: 10.1128/AAC.00361-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Helander IM, Mattila-Sandholm T. Fluorometric assessment of gram-negative bacterial permeabilization. J Appl Microbiol. 2000;88(2):213–9. doi: 10.1046/j.1365-2672.2000.00971.x [DOI] [PubMed] [Google Scholar]
  • 34.C M. Increasing the permeability of Escherichia coli using MAC13243. Sci Rep. 2017;7(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Virtanen P, Gommers R, Oliphant TE, Haberland M, Reddy T, Cournapeau D, et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat Methods. 2020;17(3):261–72. doi: 10.1038/s41592-019-0686-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.EUCAST. The European Committee on Antimicrobial Susceptibility Testing. Breakpoint tables for interpretation of MICs and zone diameters. Version 14.0. EUCAST; 2024. [Google Scholar]
  • 37.Sousa SF, Fernandes PA, Ramos MJ. Farnesyltransferase inhibitors: a detailed chemical view on an elusive biological problem. Curr Med Chem. 2008;15(15):1478–92. doi: 10.2174/092986708784638825 [DOI] [PubMed] [Google Scholar]
  • 38.Ochocki JD, Distefano MD. Prenyltransferase Inhibitors: Treating Human Ailments from Cancer to Parasitic Infections. Medchemcomm. 2013;4(3):476–92. doi: 10.1039/C2MD20299A [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kuchay S, Wang H, Marzio A, Jain K, Homer H, Fehrenbacher N, et al. GGTase3 is a newly identified geranylgeranyltransferase targeting a ubiquitin ligase. Nat Struct Mol Biol. 2019;26(7):628–36. doi: 10.1038/s41594-019-0249-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Buhaescu I, Izzedine H. Mevalonate pathway: a review of clinical and therapeutical implications. Clin Biochem. 2007;40(9–10):575–84. doi: 10.1016/j.clinbiochem.2007.03.016 [DOI] [PubMed] [Google Scholar]
  • 41.Perez-Gil J, Behrendorff J, Douw A, Vickers CE. The methylerythritol phosphate pathway as an oxidative stress sense and response system. Nat Commun. 2024;15(1):5303. doi: 10.1038/s41467-024-49483-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hoshino Y, Gaucher EA. On the Origin of Isoprenoid Biosynthesis. Mol Biol Evol. 2018;35(9):2185–97. doi: 10.1093/molbev/msy120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Heuston S, Begley M, Gahan CGM, Hill C. Isoprenoid biosynthesis in bacterial pathogens. Microbiology (Reading). 2012;158(Pt 6):1389–401. doi: 10.1099/mic.0.051599-0 [DOI] [PubMed] [Google Scholar]
  • 44.Lai YC, Peng HL, Chang HY. Identification of genes induced in vivo during Klebsiella pneumoniae CG43 infection. Infect Immun. 2001;69(11):7140–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Shin SJ, Wu C-W, Steinberg H, Talaat AM. Identification of novel virulence determinants in Mycobacterium paratuberculosis by screening a library of insertional mutants. Infect Immun. 2006;74(7):3825–33. doi: 10.1128/IAI.01742-05 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kuzuyama T, Seto H. Two distinct pathways for essential metabolic precursors for isoprenoid biosynthesis. Proc Jpn Acad Ser B Phys Biol Sci. 2012;88(3):41–52. doi: 10.2183/pjab.88.41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Palsuledesai CC, Distefano MD. Protein prenylation: enzymes, therapeutics, and biotechnology applications. ACS Chem Biol. 2015;10(1):51–62. doi: 10.1021/cb500791f [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Macek B, Forchhammer K, Hardouin J, Weber-Ban E, Grangeasse C, Mijakovic I. Protein post-translational modifications in bacteria. Nat Rev Microbiol. 2019;17(11):651–64. doi: 10.1038/s41579-019-0243-0 [DOI] [PubMed] [Google Scholar]
  • 49.Ma Q, et al. Post-translational Modifications in Oral Bacteria and Their Functional Impact. Front Microbiol. 2021;12:784923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Reinicke AT, Hutchinson JL, Magee AI, Mastroeni P, Trowsdale J, Kelly AP. A Salmonella typhimurium effector protein SifA is modified by host cell prenylation and S-acylation machinery. J Biol Chem. 2005;280(15):14620–7. doi: 10.1074/jbc.M500076200 [DOI] [PubMed] [Google Scholar]
  • 51.Price CTD, Al-Quadan T, Santic M, Jones SC, Abu Kwaik Y. Exploitation of conserved eukaryotic host cell farnesylation machinery by an F-box effector of Legionella pneumophila. J Exp Med. 2010;207(8):1713–26. doi: 10.1084/jem.20100771 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ivanov SS, Charron G, Hang HC, Roy CR. Lipidation by the host prenyltransferase machinery facilitates membrane localization of Legionella pneumophila effector proteins. J Biol Chem. 2010;285(45):34686–98. doi: 10.1074/jbc.M110.170746 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Marakasova ES, et al. Prenylation: from bacteria to eukaryotes. Mol Biol (Mosk). 2013;47(5):717–30. [PubMed] [Google Scholar]
  • 54.Hirooka K, Shioda S, Okada M. Identification of critical residues for the catalytic activity of ComQ, a Bacillus prenylation enzyme for quorum sensing, by using a simple bioassay system. Biosci Biotechnol Biochem. 2020;84(2):347–57. doi: 10.1080/09168451.2019.1685371 [DOI] [PubMed] [Google Scholar]
  • 55.Yang Q, Xue B, Liu F, Lu Y, Tang J, Yan M, et al. Farnesyltransferase inhibitor lonafarnib suppresses respiratory syncytial virus infection by blocking conformational change of fusion glycoprotein. Sig Transduct Target Ther. 2024;9(1). doi: 10.1038/s41392-024-01858-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Weber L, Mautner L, Hoyos M, Ehrhardt A, Baiker A, Bachmann HS. Effect of farnesyltransferase inhibitors on SARS-CoV-2. J Glob Antimicrob Resist. 2023;32:164–6. doi: 10.1016/j.jgar.2022.11.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Khan M, Irvin P, Park SB, Ivester HM, Ricardo-Lax I, Leek M, et al. Repurposing of lonafarnib as a treatment for SARS-CoV-2 infection. JCI Insight. 2025;10(1):e182704. doi: 10.1172/jci.insight.182704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Diani E, et al. Colistin: Lights and Shadows of an Older Antibiotic. Molecules. 2024;29(13). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Faleye OS, Boya BR, Lee J-H, Choi I, Lee J. Halogenated antimicrobial agents to combat drug-resistant pathogens. Pharmacol Rev. 2023;76(1). [DOI] [PubMed] [Google Scholar]
  • 60.Björkhem-Bergman L, Bergman P, Andersson J, Lindh JD. Statin treatment and mortality in bacterial infections--a systematic review and meta-analysis. PLoS One. 2010;5(5):e10702. doi: 10.1371/journal.pone.0010702 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hackam DG, Mamdani M, Li P, Redelmeier DA. Statins and sepsis in patients with cardiovascular disease: a population-based cohort analysis. Lancet. 2006;367(9508):413–8. doi: 10.1016/S0140-6736(06)68041-0 [DOI] [PubMed] [Google Scholar]
  • 62.Jerwood S, Cohen J. Unexpected antimicrobial effect of statins. J Antimicrob Chemother. 2008;61(2):362–4. doi: 10.1093/jac/dkm496 [DOI] [PubMed] [Google Scholar]
  • 63.Ko HHT, et al. Statins: antimicrobial resistance breakers or makers? PeerJ. 2017;5:e3952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Thangamani S, et al. Exploring simvastatin, an antihyperlipidemic drug, as a potential topical antibacterial agent. Sci Rep. 2015;5:16407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sirtori CR. The pharmacology of statins. Pharmacol Res. 2014;88:3–11. [DOI] [PubMed] [Google Scholar]
  • 66.Wang CH, et al. Microbial production of mevalonate. J Biotechnol. 2023;370:1–11. [DOI] [PubMed] [Google Scholar]
  • 67.Wilding EI, Kim DY, Bryant AP, Gwynn MN, Lunsford RD, McDevitt D, et al. Essentiality, expression, and characterization of the class II 3-hydroxy-3-methylglutaryl coenzyme A reductase of Staphylococcus aureus. J Bacteriol. 2000;182(18):5147–52. doi: 10.1128/JB.182.18.5147-5152.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wahlund TM, Tabita FR. The reductive tricarboxylic acid cycle of carbon dioxide assimilation: initial studies and purification of ATP-citrate lyase from the green sulfur bacterium Chlorobium tepidum. J Bacteriol. 1997;179(15):4859–67. doi: 10.1128/jb.179.15.4859-4867.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kanao T, Fukui T, Atomi H, Imanaka T. ATP-citrate lyase from the green sulfur bacterium Chlorobium limicola is a heteromeric enzyme composed of two distinct gene products. Eur J Biochem. 2001;268(6):1670–8. doi: 10.1046/j.1432-1033.2001.02034.x [DOI] [PubMed] [Google Scholar]
  • 70.Hohl RJ, Lewis KA, Cermak DM, Wiemer DF. Stereochemistry-dependent inhibition of RAS farnesylation by farnesyl phosphonic acids. Lipids. 1998;33(1):39–46. doi: 10.1007/s11745-998-0178-x [DOI] [PubMed] [Google Scholar]
  • 71.Pompliano DL, Rands E, Schaber MD, Mosser SD, Anthony NJ, Gibbs JB. Steady-state kinetic mechanism of Ras farnesyl:protein transferase. Biochemistry. 1992;31(15):3800–7. doi: 10.1021/bi00130a010 [DOI] [PubMed] [Google Scholar]
  • 72.Costa CB, Casalta-Lopes J, Andrade C, Moreira D, Oliveira A, Gonçalves AC, et al. Farnesyltransferase inhibitors: molecular evidence of therapeutic efficacy in acute lymphoblastic leukemia through cyclin D1 inhibition. Anticancer Res. 2012;32(3):831–8. [PubMed] [Google Scholar]
  • 73.Yu Z, Zhu Y, Fu J, Qiu J, Yin J. Enhanced NADH Metabolism Involves Colistin-Induced Killing of Bacillus subtilis and Paenibacillus polymyxa. Molecules. 2019;24(3):387. doi: 10.3390/molecules24030387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bremer E, Calteau A, Danchin A, Harwood C, Helmann JD, Médigue C, et al. A model industrial workhorse: Bacillus subtilis strain 168 and its genome after a quarter of a century. Microb Biotechnol. 2023;16(6):1203–31. doi: 10.1111/1751-7915.14257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Leeten K, et al. Ticagrelor alters the membrane of Staphylococcus aureus and enhances the activity of vancomycin and daptomycin without eliciting cross-resistance. mBio. 2024;15(10):e0132224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Jung D, Bachmann HS. Regulation of protein prenylation. Biomed Pharmacother. 2023;164:114915. doi: 10.1016/j.biopha.2023.114915 [DOI] [PubMed] [Google Scholar]
  • 77.Pantel L, Guérin F, Serri M, Gravey F, Houard J, Maurent K, et al. Exploring Cluster-Dependent Antibacterial Activities and Resistance Pathways of NOSO-502 and Colistin against Enterobacter cloacae Complex Species. Antimicrob Agents Chemother. 2022;66(11):e0077622. doi: 10.1128/aac.00776-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ren Y, Ren Y, Zhou Z, Guo X, Li Y, Feng L, et al. Complete genome sequence of Enterobacter cloacae subsp. cloacae type strain ATCC 13047. J Bacteriol. 2010;192(9):2463–4. doi: 10.1128/JB.00067-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ramirez D, Giron M. Enterobacter infections. In: StatPearls. Treasure Island (FL); 2025. [PubMed] [Google Scholar]
  • 80.Guérin F, Lallement C, Goudergues B, Isnard C, Sanguinetti M, Cacaci M, et al. Landscape of in vivo Fitness-Associated Genes of Enterobacter cloacae Complex. Front Microbiol. 2020;11:1609. doi: 10.3389/fmicb.2020.01609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Mange J-P, Stephan R, Borel N, Wild P, Kim KS, Pospischil A, et al. Adhesive properties of Enterobacter sakazakii to human epithelial and brain microvascular endothelial cells. BMC Microbiol. 2006;6:58. doi: 10.1186/1471-2180-6-58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Cheng P, Sun Y, Wang B, Liang S, Yang Y, Gui S, et al. Mechanism of synergistic action of colistin with resveratrol and baicalin against mcr-1-positive Escherichia coli. Biomed Pharmacother. 2024;180:117487. doi: 10.1016/j.biopha.2024.117487 [DOI] [PubMed] [Google Scholar]
  • 83.Gregoire N, et al. Clinical pharmacokinetics and pharmacodynamics of colistin. Clin Pharmacokinet. 2017;56(12):1441–60. [DOI] [PubMed] [Google Scholar]
  • 84.Lin JY, Zhu ZC, Zhu J, Chen L, Du H. Antibiotic heteroresistance in Klebsiella pneumoniae: Definition, detection methods, mechanisms, and combination therapy. Microbiol Res. 2024;283:127701. doi: 10.1016/j.micres.2024.127701 [DOI] [PubMed] [Google Scholar]
  • 85.F E, A A, R M. Antimicrobial susceptibility testing for polymyxins: challenges, issues, and recommendations. J Clin Microb. 2019;57(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Castaneda C, Meadows KL, Truax R, Morse MA, Kaufmann SH, Petros WP, et al. Phase I and pharmacokinetic study of lonafarnib, SCH 66336, using a 2-week on, 2-week off schedule in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2011;67(2):455–63. doi: 10.1007/s00280-010-1488-5 [DOI] [PubMed] [Google Scholar]
  • 87.Badgeley A, Anwar H, Modi K, Murphy P, Lakshmikuttyamma A. Effect of probiotics and gut microbiota on anti-cancer drugs: Mechanistic perspectives. Biochim Biophys Acta Rev Cancer. 2021;1875(1):188494. doi: 10.1016/j.bbcan.2020.188494 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Shengwei Sun

16 Jun 2025

In Vitro Synergy of Farnesyltransferase Inhibitors in Combination with Colistin against ESKAPE Bacteria

PLOS ONE

Dear Dr. Bachmann,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by Jul 31 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Shengwei Sun, Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. We note that the S1_File in your submission contains a copyrighted image. All PLOS content is published under the Creative Commons Attribution License (CC BY 4.0), which means that the manuscript, images, and Supporting Information files will be freely available online, and any third party is permitted to access, download, copy, distribute, and use these materials in any way, even commercially, with proper attribution. For more information, see our copyright guidelines: http://journals.plos.org/plosone/s/licenses-and-copyright.

We require you to either (1) present written permission from the copyright holder to publish these figures specifically under the CC BY 4.0 license, or (2) remove the figures from your submission:

 1. You may seek permission from the original copyright holder of the Figure in S1_File to publish the content specifically under the CC BY 4.0 license.

We recommend that you contact the original copyright holder with the Content Permission Form (http://journals.plos.org/plosone/s/file?id=7c09/content-permission-form.pdf) and the following text:

“I request permission for the open-access journal PLOS ONE to publish XXX under the Creative Commons Attribution License (CCAL) CC BY 4.0 (http://creativecommons.org/licenses/by/4.0/). Please be aware that this license allows unrestricted use and distribution, even commercially, by third parties. Please reply and provide explicit written permission to publish XXX under a CC BY license and complete the attached form.”

Please upload the completed Content Permission Form or other proof of granted permissions as an "Other" file with your submission.

In the figure caption of the copyrighted figure, please include the following text: “Reprinted from [ref] under a CC BY license, with permission from [name of publisher], original copyright [original copyright year].”

 2. If you are unable to obtain permission from the original copyright holder to publish these figures under the CC BY 4.0 license or if the copyright holder’s requirements are incompatible with the CC BY 4.0 license, please either i) remove the figure or ii) supply a replacement figure that complies with the CC BY 4.0 license. Please check copyright information on all replacement figures and update the figure caption with source information. If applicable, please specify in the figure caption text when a figure is similar but not identical to the original image and is therefore for illustrative purposes only.

3. Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

Reviewer #1: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously? -->?>

Reviewer #1: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available??>

The PLOS Data policy

Reviewer #1: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English??>

Reviewer #1: Yes

**********

Reviewer #1: The manuscript investigates the effects of FTIs and additional compounds on key pathogenic bacteria when co-administered with colistin. The potential synergistic interactions were illustrated by a modified checkerboard assay. The approach enables FTIs to exhibit antimicrobial activity against several gram-negative bacteria when co-administered with sub-inhibitory concentrations of colistin. The authors found FTIs were able to reduce colistin MICs, even in a colistin-resistant gram-negative strain, thus offering a novel strategy to combat antimicrobial resistance. However, several key issues are not clarified clearly, some modification should be made according to the following questions:

Comments:

1. Line 216, why does S. aureus MRSA have antibiotic resistance to the quantities of ampicillin, chloramphenicol, kanamycin and tetracycline, while S. aureus doesn’t?

2. Line 223, the author should provide the specific value of the sub-inhibitory concentration of colistin.

3. In the “Growth kinetics” section, the author only conducted the growth kinetics for S. aureus. Why did the authors not monitor the dynamic changes in the combined inhibitory effects of FTIs and colistin on E. cloacae growth? Time-kill assays should be included to evaluate the antibacterial effect.

4. Line 382, the author should discuss the lack of activity of the peptidomimetic FTIs, B581 and FTI-277 against gram-positive bacteria. Why the antimicrobial effects of peptidomimetic FTIs lower than non-peptidomimetic FTIs?

5. Line 438-440 and figure 8, the author only speculated the mechanism of the synergistic effects on gram-negative bacteria with the combined use of colistin and FTIs by disrupting the OM’s integrity and increasing permeability. The conclusion is not supported by any data. To further strengthen the conclusions, the authors may consider performing outer membrane permeability assay to confirm.

6. Line 476-479, “…colistin-resistant strains, including those harboring mobile colistin resistance genes “, “this approach could target bacteria with a strong propensity for heteroresistance to colistin and other polymyxins”, please give examples to illustrate which bacteria are included respectively.

7. Line 481-483, could you give more examples/details about how the FTIs affect the heteroresistance in gram-negative pathogens bacteria with or without colistin?

**********

what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy

Reviewer #1: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org

PLoS One. 2025 Sep 5;20(9):e0331440. doi: 10.1371/journal.pone.0331440.r002

Author response to Decision Letter 1


30 Jul 2025

Dear Editor, dear Reviewer,

We would like to sincerely thank you for your thorough evaluation of our manuscript and the opportunity to contribute to PLOS ONE. We greatly appreciate the constructive feedback and the chance to further improve our work in response to your valuable comments.

We have carefully reviewed all submitted files and, to the best of our knowledge, ensured that they now fully comply with PLOS ONE's style and file naming requirements. Concerning the use of the image in the S1_File, we contacted the Tecan Deutschland GmbH, which provides both the device and the associated software shown in the screenshot. We received written confirmation that the reported screenshot does not constitute a copyright issue and can be used within the scope of this publication without permission. The relevant correspondence is provided as a supporting document.

We now respond point-by-point to the reviewer’s comments below.

1. Line 216, why does S. aureus MRSA have antibiotic resistance to the quantities of ampicillin, chloramphenicol, kanamycin and tetracycline, while S. aureus doesn’t?

We agree that this aspect needed clarification. The MRSA strain used in our study, Staphylococcus aureus ATCC 33592, is mecA-positive (PMID: 19752281), whereas S. aureus ATCC 25923 is mecA-negative and widely used as a quality control strain (PMID: 25377701). The methicillin-sensitive strain is markedly more susceptible to the antibiotics tested compared to the methicillin-resistant one. It is well established that methicillin resistance in S. aureus is frequently associated with cross-resistances, and thus often coincides with multidrug resistance (PMID: 32257966). To demonstrate that established antibiotics can still be effective against this MRSA strain, we additionally included gentamicin and vancomycin in our control experiments, as shown in Fig 3.

2. Line 223, the author should provide the specific value of the sub-inhibitory concentration of colistin.

The exact concentrations of colistin used in the combination experiments are already detailed in the Materials and Methods section under the description of the checkerboard assay, and are also indicated in the respective figures. Nevertheless, in accordance with the reviewer’s recommendation, we have now included the concentration range directly in the Results section for clarity (Line 248-250).

3. In the “Growth kinetics” section, the author only conducted the growth kinetics for S. aureus. Why did the authors not monitor the dynamic changes in the combined inhibitory effects of FTIs and colistin on E. cloacae growth? Time-kill assays should be included to evaluate the antibacterial effect.

This is a valid point, and we have now extended our analysis by conducting additional growth kinetics for E. cloacae, specifically focusing on the combined application of colistin and FTIs to illustrate potential synergistic effects. Furthermore, we have implemented time-kill assays for both S. aureus and E. cloacae to more thoroughly assess the bactericidal activity and the dynamic interplay between colistin and FTIs over time. These new experiments have been incorporated into the revised manuscript (Line 33, 185-192, 210-213, 340, and 351-390).

4. Line 382, the author should discuss the lack of activity of the peptidomimetic FTIs, B581 and FTI-277 against gram-positive bacteria. Why the antimicrobial effects of peptidomimetic FTIs lower than non-peptidomimetic FTIs?

The differences in antimicrobial activity between the tested FTIs likely result from distinct chemical properties. Lonafarnib and tipifarnib, which affect gram-positive and gram-negative bacteria, both carry halogen substituents (bromine and chlorine, respectively). Halogens are known to be important for the antibiotic activity of different classes of antibiotics, like chloramphenicol, tetracyclines, and fluoroquinolones. Faleye et al. (2023; PMID: 37845080) emphasized that halogenated compounds often show increased affinity for gram-positive bacteria. Similarly, Sanabria-Rios et al. (2022; PMID: 36118101) demonstrated brominated fatty acids to be effective against MRSA strains. These findings may explain the higher activity of lonafarnib and tipifarnib. In contrast, the peptidomimetic FTIs B581 and FTI-277 contain polar thiol groups, which may impair membrane permeability. We have added an explanation of the chemical structures and properties of the molecules to the manuscript (Line 471-475). Further investigations into membrane integrity and compound uptake will help clarify the role of these functional groups in antimicrobial activity.

5. Line 438-440 and figure 8, the author only speculated the mechanism of the synergistic effects on gram-negative bacteria with the combined use of colistin and FTIs by disrupting the OM’s integrity and increasing permeability. The conclusion is not supported by any data. To further strengthen the conclusions, the authors may consider performing outer membrane permeability assay to confirm.

To address this concern, we conducted outer membrane permeability assays using the fluorescent probe N-Phenyl-1-naphthylamine (NPN) for E. coli as a model strain and E. cloacae, a colistin-resistant isolate, as representative gram-negative bacteria. The results confirmed that sub-inhibitory concentrations of colistin were sufficient to increase outer membrane permeability without affecting viability. These new data have been included in the revised manuscript (Line 194-204, 392-415).

6. Line 476-479, “…colistin-resistant strains, including those harboring mobile colistin resistance genes “, “this approach could target bacteria with a strong propensity for heteroresistance to colistin and other polymyxins”, please give examples to illustrate which bacteria are included respectively.

As we were able to demonstrate a significant antimicrobial effect of FTIs against gram-negative bacteria when co-administered with sub-inhibitory concentrations of colistin, the required colistin concentration was notably reduced. This finding suggests that even colistin-resistant bacteria might be susceptible to such combination treatments. Notably, one of the tested strains, E. cloacae ATCC 13047, is known to be highly resistant to colistin (PMID: 20207761). Members of the Enterobacter cloacae complex (ECC) are not only important nosocomial pathogens but are also well documented for their high potential to exhibit colistin heteroresistance (PMID: 36627272). Furthermore, plasmid-mediated colistin resistance genes (mcr) have been identified across several gram-negative ESKAPE organisms, including E. coli (PMID: 30305321), K. pneumoniae (PMID: 34946038), and A. baumannii (PMID: 30936095). While we did not include mcr-positive isolates in our study, future work will investigate whether FTIs may help restore susceptibility in strains carrying mcr-mediated colistin resistance. Notably, previous studies have shown that mcr-mediated colistin resistance can be overcome by using rationally designed combination therapies (PMID: 29386620). These findings underscore the potential of similar approaches – such as combining FTIs with sub-inhibitory colistin – to restore susceptibility in resistant gram-negative pathogens.

7. Line 481-483, could you give more examples/details about how the FTIs affect the heteroresistance in gram-negative pathogens bacteria with or without colistin?

This point raises an important aspect of our findings. When used alone, FTIs showed no impact on most gram-negative bacteria, clearly indicating resistance rather than heteroresistance. In our study, we mentioned the possibility that FTIs in combination with colistin potentially influence the formation of heteroresistant subcolonies. For instance, lonafarnib, when combined with sub-inhibitory colistin, did not consistently suppress bacterial growth across the triplicate near the MIC threshold in the K. pneumoniae checkerboard assay, which supports the hypothesis of heteroresistant escape events. However, this remains a hypothesis. We have therefore deleted the word 'heteroresistant' from the Results section and now describe the formation of subcolonies only.

However, we assume that the observed inconsistencies in growth inhibition patterns, particularly near the MIC threshold, are characteristic of heteroresistance, where subpopulations can transiently survive otherwise inhibitory concentrations (PMID: 25567227). We therefore proceed to discuss this hypothesis in greater detail in the Discussion (Line 574-576).

Attachment

Submitted filename: Response_to_Reviewers.docx

pone.0331440.s013.docx (15.4KB, docx)

Decision Letter 1

Shengwei Sun

18 Aug 2025

In Vitro Synergy of Farnesyltransferase Inhibitors in Combination with Colistin against ESKAPE Bacteria

PONE-D-25-17781R1

Dear Dr. Bachmann,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager®  and clicking the ‘Update My Information' link at the top of the page. For questions related to billing, please contact billing support .

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Shengwei Sun, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Acceptance letter

Shengwei Sun

PONE-D-25-17781R1

PLOS ONE

Dear Dr. Bachmann,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

You will receive further instructions from the production team, including instructions on how to review your proof when it is ready. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few days to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

You will receive an invoice from PLOS for your publication fee after your manuscript has reached the completed accept phase. If you receive an email requesting payment before acceptance or for any other service, this may be a phishing scheme. Learn how to identify phishing emails and protect your accounts at https://explore.plos.org/phishing.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Shengwei Sun

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 File. Additional information and controls.

    The file provides supporting information on the Tecan device and the program used for fluorescence measurements, as well as details of the conducted controls.

    (DOCX)

    pone.0331440.s001.docx (1.2MB, docx)
    S1 Fig. RFI values for A. baumannii.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for A. baumannii. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s002.tif (468.3KB, tif)
    S2 Fig. RFI values for B. subtilis.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for B. subtilis. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s003.tif (464.7KB, tif)
    S3 Fig. RFI values for E. cloacae.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for E. cloacae. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s004.tif (451.3KB, tif)
    S4 Fig. RFI values for E. faecium.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for E. faecium Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s005.tif (477.9KB, tif)
    S5 Fig. RFI values for E. coli.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for E. coli. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s006.tif (462.1KB, tif)
    S6 Fig. RFI values for K. pneumoniae.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for K. pneumoniae. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s007.tif (468.4KB, tif)
    S7 Fig. RFI values for MRSA.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for MRSA. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s008.tif (459.3KB, tif)
    S8 Fig. RFI values for P. paraeruginosa.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for P. paraeruginosa. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s009.tif (467.4KB, tif)
    S9 Fig. RFI values for S. aureus.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for S. aureus. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s010.tif (461.6KB, tif)
    S10 Fig. RFI values for S. epidermidis.

    Relative fluorescence intensity (RFI) values for all checkerboard assays with colistin and the six tested substances for S. epidermidis. Minimum bactericidal concentrations (MBCs) are indicated in bold and italic. All values were normalized to the fluorescence of the corresponding growth control to ensure comparability across plates. All checkerboard assays were conducted in triplicate.

    (TIF)

    pone.0331440.s011.tif (466.8KB, tif)
    Attachment

    Submitted filename: Response_to_Reviewers.docx

    pone.0331440.s013.docx (15.4KB, docx)

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting Information files.


    Articles from PLOS One are provided here courtesy of PLOS

    RESOURCES