Skip to main content
Journal of Antimicrobial Chemotherapy logoLink to Journal of Antimicrobial Chemotherapy
. 2022 Dec 7;78(2):397–410. doi: 10.1093/jac/dkac402

Oral ciprofloxacin activity against ceftriaxone-resistant Escherichia coli in an in vitro bladder infection model

Iain J Abbott 1,, Elke van Gorp 2, Hugh Cottingham 3, Nenad Macesic 4, Steven C Wallis 5, Jason A Roberts 6,7,8, Joseph Meletiadis 9,b, Anton Y Peleg 10,11,b
PMCID: PMC9890216  PMID: 36473954

Abstract

Objectives

Pharmacodynamic profiling of oral ciprofloxacin dosing for urinary tract infections caused by ceftriaxone-resistant Escherichia coli isolates with ciprofloxacin MIC ≥ 0.25 mg/L.

Background

Urine-specific breakpoints for ciprofloxacin do not exist. However, high urinary concentrations may promote efficacy in isolates with low-level resistance.

Methods

Ceftriaxone-resistant E. coli urinary isolates were screened for ciprofloxacin susceptibility. Fifteen representative strains were selected and tested using a dynamic bladder infection model. Oral ciprofloxacin dosing was simulated over 3 days (250 mg daily, 500 mg daily, 250 mg 12 hourly, 500 mg 12 hourly and 750 mg 12 hourly). The model was run for 96 h. Primary endpoint was change in bacterial density at 72 h. Secondary endpoints were follow-up change in bacterial density at 96 h and area-under-bacterial-kill-curve. Bacterial response was related to exposure (AUC0–24/MIC; Cmax/MIC). PTA was determined using Monte-Carlo simulation.

Results

Ninety-three clinical isolates demonstrated a trimodal ciprofloxacin MIC distribution (modal MICs at 0.016, 0.25 and 32 mg/L). Fifteen selected clinical isolates (ciprofloxacin MIC 0.25–512 mg/L) had a broad range of quinolone-resistance genes. Following ciprofloxacin exposure, E. coli ATCC 25922 (MIC 0.008 mg/L) was killed in all dosing experiments. Six isolates (MIC ≥ 16 mg/L) regrew in all experiments. Remaining isolates (MIC 0.25–8 mg/L) regrew variably after an initial period of killing, depending on simulated ciprofloxacin dose. A >95% PTA, using AUC0–24/MIC targets, supported 250 mg 12 hourly for susceptible isolates (MIC ≤ 0.25 mg/L). For isolates with MIC ≤ 1 mg/L, 750 mg 12 hourly promoted 3 log10 kill at the end of treatment (72 h), 1 log10 kill at follow-up (96 h) and 90% maximal activity (AUBKC0–96).

Conclusions

Bladder infection modelling supports oral ciprofloxacin activity against E. coli with low-level resistance (ciprofloxacin MIC ≤ 1 mg/L) when using high dose therapy (750 mg 12 hourly).

Introduction

Urinary tract infections (UTIs) are extremely common.1 Severe infection accounts for 25% of emergency sepsis cases, with a 6.2% in-hospital mortality and 8.6% ICU mortality.2Escherichia coli remains the most common uropathogen. Antimicrobial resistance (AMR) and the global spread of the MDR strain E. coli ST131 and, more recently, E. coli ST1193, greatly limits empirical therapy in serious infections.3–7 Rates of resistance have progressively increased since 2007, with an 8-fold rise in resistance genes and increased numbers of attributable deaths and disability-adjusted life years.8–10 AMR in UTIs is the one of the leading infectious syndromes contributing to the global burden of deaths associated with resistance.11 Globally in 2019, UTIs caused by E. coli with antimicrobial resistance resulted in >26 000 deaths directly attributable to AMR and >100 000 deaths related to AMR.12 This was predominantly in E. coli with resistance to third-generation cephalosporins and fluoroquinolones.

Oral ciprofloxacin has excellent bioavailability and tissue penetration and achieves high concentrations in urine compared with plasma (Table 1). It has been well studied in the treatment of infections arising from the urinary tract. The US FDA and the EMA, however, provide black box warnings about the potential harms with the use of fluoroquinolones, including tendinopathy, aortic ruptures or tears and CNS effects.23,24 As such, ciprofloxacin is not recommended for routine use in mild or recurrent UTIs. Alternative antibiotics with a narrower-spectrum, better side-effect profile, and less microbiome disruption are recommended to be used whenever possible, such as nitrofurantoin, fosfomycin, pivmecillinam and co-trimoxazole. The EUCAST 2022 breakpoints for ciprofloxacin are ‘susceptible, standard dosing regimen’ (S) MIC ≤ 0.25, and ‘resistant’ (R) MIC > 0.5 mg/L. An MIC measurement of 0.5 mg/L is designated as an area of technical uncertainty (ATU), dealt by the laboratory (depending on type of sample, number of alternative agents, severity of infection, consultation with clinical colleagues). Breakpoints are based on standard oral dosing of 500 mg 12 hourly, with high dose defined as 750 mg 12 hourly.25 There are no UTI-specific breakpoints. CLSI 2022 breakpoints for ciprofloxacin are ‘susceptible’ (S) MIC ≤ 0.25 mg/L, ‘intermediate’ (I) MIC = 0.5 mg/L^, ‘resistant’ (R) MIC ≥ 1 mg/L, where ‘^’ indicates the potential to concentrate in the urine. Breakpoints are also based on oral 500 mg 12 hourly.26 The decision to report ‘I^’ is made by each laboratory based on institution-specific guidelines and in consultation with medical personnel. A lower dose of ciprofloxacin of 250 mg 12 hourly is commonly used for uncomplicated UTIs.27

Table 1.

Oral ciprofloxacin urinary pharmacokinetics

Year Subjects n Dose Time (h) Mean (mg/L) Min Max % CV Recovery in urine Method Ref
1984 Healthy volunteers (all male) 12 250 mg q12; 7 days (13 doses) 0–2 a 72 840 HPLC 13
6–12 44.7 ± 25 to 68.7 ± 45 56 to 77%
1984 Healthy volunteers (all male) 9 250, 500, 750 mg q12; 7 days 0–2 (250 mg)
2–4
4–8
8–12
205 to 261
147 to 229
90 to 101
32 to 34
0–12 h (250 mg): 41% ± 7% to 47% ± 19% Bioassayb 14
0–2 (500 mg)
2–4
4–8
8–12
255 to 518
321 to 448
117 to 199
26 to 82
0–12 h (500 mg): 33% ± 9% to 37% ± 10%
0–2 (750 mg)
2–4
4–8
8–12
243 to 846
544 to 704
169 to 360
55 to 151
0–12 h (750 mg): 31% ± 7% to 38% ± 8%
1985 Healthy volunteers (50% female) 12 50 mg, 100 mg, 750 mg; single dose 0–3, 3–6, 6–12 & 12–24 a 0–24 h (50 mg): 36% ± 10% Bioassayc 15
0–24 h (100 mg): 35% ± 8%
0–24 h (750 mg): 33% ± 5%
1985 Healthy volunteers (50% female) 10 250 mg q12; 4 days (7 doses) d a 0–12 h: 38% ± 6% to 46% ± 6% (bioassay); 26% ± 5% to 30% ± 9% (HPLC) Bioassaye and HPLCf 16
1985 Healthy volunteers (all male) 18 500 mg; single dose 0–2
2–4
4–8
8–12
464
304
189
111
28
60
31
19
1847
1219
731
128
98%
95%
93%
25%
0–12 h: 45% ± 12%
0–24 h: 51% ± 12.2%
HPLC 17
1986 Healthy volunteers (all male) 12 250, 500, 750, 1000 mg; single dose 0–2 (250 mg)
2–4
4–8
8–12
190g
180
90
40
0–48 h (250 mg): 36% ± 12% Bioassayb 18
0–2 (500 mg)
2–4
4–8
8–12
340g
400
220
120
0–48 h (500 mg): 44% ± 7.5%
0–2 (750 mg)
2–4
4–8
8–12
460g
530
280
200
0–48 h (750 mg): 40% ± 12.2%
0–2 (1000 mg)
2–4
4–8
8–12
390g
400
240
130
0–48 h (1000 mg): 29% ± 10%
1994 Healthy volunteers (all male) 6 750 mg; single dose 0–4, 4–8, 8–12, 12–24 a,h 0–12 h: 41% (bioassay); 36% (HPLC) Bioassaye and HPLCf 19
2003 Healthy volunteers (50% female) 12 500 mg; single dose 0–6
6–12
407
47
23
19
733
76
44%
30%
0–12 h: 37% ± 10% HPLC 20
2004 Healthy volunteers (50% female) 12 500 mg; single dose 0–6
6–12
368
77
100
30
1000
179
61%
48%
0–12 h: 32% ± 10% HPLC 21
2006 Healthy volunteers (50% female) 14 500 mg; single dose 0–6
6–12
268
60
130
25
967
76
78%
21%
0–12 h: 34% ± 8% HPLC 22

Average urinary concentrations not provided.

Klebsiella pneumoniae ATCC 10031.

K. pneumoniae ATCC 10031 for concentrations <0.15 mg/L and Bacillus subtilis ATCC 6633 for concentrations >0.15 mg/L.

Urine collected at seven time intervals after the first and seventh doses. Average urinary concentrations not provided.

E. coli 4004.

Bioassay results higher than HPLC, supporting the assumption that microbiologically active metabolites are excreted renally.

Average urinary concentrations were only presented in graphical form; measurements are approximated from the figure.

Authors state that urinary concentrations were >26 mg/L from all 12–24 urine samples.

A recent systematic review highlights the global rising rates of fluoroquinolone-resistant E. coli in community-acquired uncomplicated UTIs in women, with resistance rate rising from 0.5% to 15.3% in the UK, 8.7% to 15.1% in Germany, 22.9% to 30.8% in Spain, 4% to 12% in North America, and 25% to >40% in Asia.28 Given high urinary concentrations achieved after oral dosing, it is hypothesized that urinary isolates categorized as ciprofloxacin resistant could still be effectively treated with high-dose ciprofloxacin. We have performed pharmacodynamic profiling of different oral ciprofloxacin dosing schedules within a dynamic bladder infection in vitro model to assess the applicability of UTI-specific ciprofloxacin breakpoints against ceftriaxone-resistant E. coli urinary isolates.

Methods

Media

Cation-adjusted Mueller-Hinton II agar (MHA, BD, USA), CAMHB (BD), pooled human urine and synthetic human urine (SHU) were used. Human urine was collected and pooled from healthy female volunteers and filter sterilized (Ethics Committee approval: Project no. 27033). SHU was modified (mSHU) from a previous study29 with addition of 0.1% v/v yeast extract (stock solution 10% w/v) to best match E. coli growth in urine (Table S1 and Figure S1, available as Supplementary data at JAC Online). SHU and mSHU were adjusted to pH 5.6.

E. coli isolates and susceptibility testing

Clinical, non-duplicate E. coli isolates from a urinary source that were ceftriaxone resistant were selected from a surveillance collection at a tertiary acute care hospital (Ethics Committee approval: Project no. 533/16). Isolates underwent ciprofloxacin susceptibility testing by broth microdilution (BMD).30 Fifteen clinical isolates, and E. coli ATCC 25922, were selected to reflect a range of ciprofloxacin MIC values. Ciprofloxacin MICs were repeated in triplicate by BMD in CAMHB and mSHU, and tested in pooled human urine as a single replicate. WGS was performed determining ST, phylogenetic relatedness, quinolone-resistance determinants and β-lactamase genes (see Supplementary Methods).

Bladder infection in vitro model

A multicompartment infection model applying a continuous dilution system was used with mSHU as the liquid medium (Figure 1).31 Sixteen bladder compartments were run in parallel. Medium was run at a continuous flow rate of 400 mL/h from fresh media reservoirs into the ‘intestinal’ compartment containing ciprofloxacin (Aspen Pharmacare Australia, 200 mg/100 mL; volume 300–900 mL), and then into the ‘circulatory’ compartment (volume 1450 mL). The volumes of these two compartments were kept static for the duration of each experiment. Medium flow into each individual bladder compartment was 25 mL/h. Normal human urodynamics was simulated. Volume in each bladder increased over time prior to an intermittent voiding schedule that reduced the volume to a residual 5 mL. First void was 2 h after starting and continued 4 hourly thereafter. Each bladder was inoculated with an E. coli isolate with 10 mL of 106 cfu/mL, providing a total bacterial count equivalent to human UTIs (i.e. ≥105 cfu/mL in 200 mL void). Ciprofloxacin MICs were rechecked from the starting inoculum and if an isolate regrew at the completion of an experiment.

Figure 1.

Figure 1.

Bladder infection model. In vitro model set-up of the dynamic multicompartment dilution model used for the simulation urinary ciprofloxacin exposure following oral dosing. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

Quantification of bacterial growth

Bacterial density (cfu/mL) was first assessed under drug-free conditions over 24 h. During ciprofloxacin exposure over 3 days, bacterial density was measured at 0, 6, 24, 30, 48, 54 h and at the end of treatment (72 h). A follow-up bacterial density was measured at 96 h. Pharmacodynamic (PD) samples were collected directly from each bladder compartment. To negate antibiotic carry-over, a centrifuge/wash process was performed twice, reducing ciprofloxacin concentrations 100-fold. Bacterial loss was minimal (<0.1 log10 cfu/mL). Samples were serially diluted and 20 µL from each dilution plated onto MHA. Emergence of resistance was identified by plating in parallel onto MHA with 2 mg/L and 128 mg/L ciprofloxacin (every 24 h). Plates were incubated aerobically, 35°C ± 1°C for 16–20 h. Plates with ciprofloxacin were reincubated for an additional 24 h. Limit of detection was 50 cfu/mL.

Simulated ciprofloxacin urinary exposure

Human urinary ciprofloxacin concentrations after oral dosing (Table 1) were used as targets for the in vitro model.13–22 Drug distribution equations were used to inform in vitro flow rate, volumes and ciprofloxacin dosage to achieve the target concentrations and a calculated AUC0–24.32 Dosing schedules were administered as a 3 day course. Target exposures [peak concentration (Cmax], trough concentration (Cmin) and calculated AUC0–24 on the third day of treatment were as follows: 250 mg daily (Cmax 220 mg/L, Cmin 1 mg/L, AUC0–24 1444 mg·h/L), 500 mg daily (Cmax 398 mg/L, Cmin 3 mg/L, AUC0–24 2969 mg·h/L), 250 mg 12 hourly (Cmax 232 mg/L, Cmin 30 mg/L, AUC0–24 2880 mg·h/L), 500 mg 12 hourly (Cmax 426 mg/L, Cmin 79 mg/L, AUC0–24 5937 mg·h/L), 750 mg 12 hourly (Cmax 579 mg/L, Cmin 154 mg/L, AUC0–24 8969 mg·h/L). Greater than 90% of each administered dose was excreted by 12 h.

Measurement of ciprofloxacin concentrations

Representative pharmacokinetic (PK) samples were collected from three bladder compartments at Cmax and Cmin each day of dosing, and at the completion of each experiment. All 16 bladder compartments were sampled at the Cmax on the third day. Interday and intercompartment variability was assessed by the average relative standard deviation. Samples were filtered, stored at −80°C and batched for testing. Ciprofloxacin concentrations were measured by a UHPLC with fluorometric detection (UHPLC-Fl) method on a Nexera2 liquid chromatograph connected to an RF-20Axs fluorescence detector (Shimadzu, Kyoto, Japan). Calibration range was 0.1 to 1000 mg/L. The precision (6.9%, 6.4%, 4.3% and 1.4%) and accuracy (0.9%, 5.0%, 10.%5 and −2.2% at 3, 30, 300 and 1000 mg/L) met FDA guidance.33 Ciprofloxacin was stable when incubated in SHU, with 0.2%, 1.9% and 4.6% reduction in the measured concentration at 24, 48 and 72 h, respectively. Linear regression and Bland–Altman plots quantified the accuracy and bias of the measured concentrations compared to target.

PK/PD analysis

Primary endpoint was end-of-treatment (72 h) change in bacterial density. Secondary endpoints were change in bacterial density at follow-up (96 h), and total bacterial response measured by the area under the bacterial kill curve (AUBKC0–96). The relationship between ciprofloxacin exposure (AUC0–24/MIC and Cmax/MIC) and bacterial response was assessed using an Emax non-linear regression model. No constraints were applied, unless indicated. Goodness of fit was assessed by visual inspection, residuals analysis and R2. Exposures required for stasis, 1, 2 and 3 log10 kill, and 50% (EI50) and 90% (EI90) of the maximum effect was determined. This analysis was repeated using MIC measurements by BMD in mSHU. Monte Carlo simulations (MCS), using the ‘normal random number generator’ for 5000 patients (Microsoft® Excel for Mac, v16.63) was used to determine PTA for each ciprofloxacin dosing regimen, with ±50% allowance for variability in human urinary concentrations. Protein binding in urine is minimal. All data were analysed using GraphPad Prism (version 9.4.1 macOS).

Results

E. coli isolates

Ninety-three ceftriaxone-resistant E. coli clinical urinary isolates were selected for ciprofloxacin MIC testing. A trimodal MIC distribution was observed with modal MICs at 0.016, 0.25 and 32 mg/L (Figure 2). Most isolates (86%) demonstrated a ciprofloxacin MIC higher than the epidemiological cut-off (ECOFF) of 0.064 mg/L.34 Clinical isolates selected for additional testing had ciprofloxacin MICs ranging from 0.25 to 512 mg/L (Table 2). Compared with standard testing in CAMHB, ciprofloxacin MIC measurements were, on average, 4 (±1) log2 dilutions higher when tested in mSHU and in pooled human urine. Isolates reflected a diverse range of STs and were not members of a transmission cluster (>45 SNPs between all five E. coli ST131). All isolates with ciprofloxacin MIC ≥ 4 mg/L had parC S80I mutation (Table 2). Sequence reads were deposited in the NCBI Sequence Read Archive (Table S2).

Figure 2.

Figure 2.

Ciprofloxacin MIC distribution of ceftriaxone-resistant E. coli urinary isolates. E. coli urinary isolates (n = 93). MIC testing performed by BMD. WT isolates defined by MIC ≤ 0.064 mg/L. Susceptibility categories as per EUCAST 2022 breakpoint table. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

Table 2.

Ceftriaxone-resistant E. coli isolates tested in the bladder infection model

Isolate # Ciprofloxacin MIC, mg/L (range) ST QRDRs Plasmid-mediated quinolone resistance Multidrug- efflux-pump regulator β-Lactamase
CAMHB mSHU Urine
25922 0.008 (0.008) 0.125 (0.125) 0.0625 73
057 0.25 (0.25) 8 (8) 4 131 gyrA_S83L
parE_I529L
CTX-M-27
017 0.5 (0.25–0.5) 8 (8) 8 219 qnrS1 CTX-M-15
014 0.5 (0.25–0.5) 8 (8) 4 38 gyrA_S83L CTX-M-14
015 0.5 (0.5) 8 (8) 8 131 gyrA_S83L
parC_A108V
parE_I529L
CTX-M-27
016 0.5 (0.5) 8 (8) 8 131 gyrA_S83L
parE_I529L parE_S458A
CTX-M-14
019 1 (1) 16 (16) 8 10320 gyrA_S83L
parE_S458A
CTX-M-15
114 4 (4–8) 256 (256) 64 2599 gyrA_S83L gyrA_D87N
parC_S80I
CTX-M-15
132 8 (4–8) 64 (64) 128 648 gyrA_S83L
parC_S80I
parE_S458A
qnrS1 CTX-M-65
104 8 (8) 256 (256) 256 95 gyrA_S83L gyrA_D87N
parC_S80I
marR_S3N CTX-M-55
093 16 (16) 512 (512) 512 1193 gyrA_S83L gyrA_D87N
parC_S80I
parE_L416F
qnrS1a marR_S3N CTX-M-14
124 32 (32) 512 (512) >512 131 gyrA_S83L gyrA_D87N
parC_S80I parC_E84V
parE_I529L
CTX-M-15
096 32 (32) 512 (512) >512 410 gyrA_S83L gyrA_D87N
parC_S80I
parE_S458A
CTX-M-15 CMY-42
127 64 (64) >512 (>512) >512 457 gyrA_S83L gyrA_D87Y
parC_S80I
parE_S458A
CMY-2
139 128 (128) 512 (512) >512 131 gyrA_S83L gyrA_D87N
parC_S80I parC_E84V
parE_I529L
CTX-M-15
087 512 (512) >512 (>512) >512 1193 gyrA_S83L gyrA_D87N
parC_S80I
parE_L416F
qepA8b marR_S3N CTX-M-15, CMY-2

Ciprofloxacin MIC testing in CAMHB and mSHU was performed in triplicate by BMD using ciprofloxacin HCl (Sigma–Aldrich USA, PHR1044); median (range) reported. Testing in pooled human urine was performed as a single replicate using the parental formulation of ciprofloxacin (Aspen Pharmacare Australia). Testing performed in CAMHB and mSHU. QRDRs include mutations DNA gyrase (gyr) and topoisomerase IV (par) genes.

Partial genomic sequence obtained (104/657 bases missing from assembly).

Partial genomic sequence obtained (286/1542 bases missing from assembly). The ATCC 25922 strain matched the publicly available sequence.

Simulated ciprofloxacin exposure in the bladder infection model

Ciprofloxacin concentrations closely matched target values with the linear regression slope 0.90 (R2 = 0.9758) and bias −1.9% (95% CI: −35.2% to 31.5%) (Figures S2 and S3). Intercompartment variation in Cmax across all 16 bladders on the third day treatment was 3.3% ± 0.8%. Interday variation in Cmax in the representative sampled bladders was 7.6% ± 5.9%. All ciprofloxacin concentrations measured at follow-up (96 h) were <0.27 mg/L.

Post-exposure growth response in the bladder infection model

E. coli ATCC 25922 (ciprofloxacin MIC 0.008 mg/L) was eradicated in all dosing regimens. The six clinical isolates with ciprofloxacin MIC ≥ 16 mg/L had near maximal regrowth at 72 h (>1.9 Δlog10 cfu/mL) in all dosing regimens (Table 3 and Figure 3). For the remaining nine clinical isolates (ciprofloxacin MIC 0.25–8 mg/L), after an initial period of killing, regrowth at 72 h (>0 Δlog10 cfu/mL) was: three isolates (114, 132, 104 with MICs 4–8 mg/L) after 250 mg and 500 mg daily; one isolate (104 with MIC 8 mg/L) after 250 mg and 500 mg 12 hourly; none after 750 mg 12 hourly. At follow-up (96 h) regrowth in these nine isolates was: seven isolates (057, 017, 016, 019, 114, 132, 104 with MICs 0.5–8 mg/L) after 250 mg daily; five isolates (014, 016, 114, 132, 104 with MICs 0.5–8 mg/L) after 500 mg daily; three isolates (114, 132, 104) after 250 mg and 500 mg 12 hourly; one isolate (114 with MIC 4 mg/L) after 750 mg 12 hourly.

Table 3.

Change in bacterial density at 72 and 96 h following oral ciprofloxacin dosing simulations

Isolate # CIP MIC (mg/L) End of treatment (72 h Δcfu/mL) Follow-up (96 h Δcfu/mL)
250 mg daily 500 mg daily 250 mg q12 500 mg q12 750 mg q12 250 mg daily 500 mg daily 250 mg q12 500 mg q12 750 mg q12
25922 0.008
057 0.25 1.35 −1.52
017 0.5 1.74
014 0.5 −0.43 1.17
015 0.5
016 0.5 −0.46 1.96 1.08
019 1 −2.83 1.57 −3.29
114 4 2.27 0.86 −0.68 −0.50 2.18 1.79 1.82 1.94 1.26
132 8 1.92 1.47 −3.69 −0.48 2.00 1.69 1.44 2.12
104 8 2.19 2.05 0.41 1.51 2.13 1.87 1.82 2.04
093 16 2.08 2.23 2.48 2.95 2.51 1.98 2.27 2.12 2.44 2.25
124 32 3.17 2.20 2.68 3.02 1.98 2.65 2.06 2.28 3.11 1.98
096 32 2.57 2.21 2.64 3.81 2.56 2.82 1.81 1.43 2.74 2.15
127 64 1.89 1.96 2.29 2.10 2.10 2.23 1.80 1.94 2.00 2.22
139 128 2.09 1.94 2.74 3.03 1.98 2.00 1.96 1.74 2.26 2.18
087 512 1.96 1.77 2.51 2.14 2.10 1.91 1.88 1.81 2.01 2.18

CIP, ciprofloxacin. CIP MIC as determined by BMD in CAMHB. — indicates that growth was not detected. Limit of detection was considered to be 50 cfu/mL.

Figure 3.

Figure 3.

Growth response following urinary ciprofloxacin exposure over 96 h incubation in the bladder infection model following the five dosing regimens (a–e). Sixteen E. coli isolates tested, each with a unique symbol. Limit of detection was considered to be 50 cfu/mL. Circled times on the x-axis indicate the time when repeat doses of ciprofloxacin were added to the in vitro model. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

Emergence of resistance

Ciprofloxacin MIC testing of post-exposure growth from drug-free MHA did not identify any isolate with >2 log2 dilution rise MIC compared with the starting inoculum (Table S3). Only isolate 019 (MIC 0.5 to 2 mg/L after 250 mg 12 hourly) and isolate 127 (MIC 32 to 128 mg/L after 750 mg 12 hourly) had appreciable MIC rises. Emergence of resistance was also not detected on the bacterial density measurements on MHA supplemented with 2 and 128 mg/L ciprofloxacin. MHA with 2 mg/L ciprofloxacin suppressed all isolates with ciprofloxacin MIC < 2 mg/L. MHA with 128 mg/L ciprofloxacin suppressed all isolates with ciprofloxacin MIC 2–64 mg/L, except 127 (ciprofloxacin MIC 64 mg/L), which had low-level growth at 96 h (2–3 log10 cfu/mL) following 250 mg daily, 250 mg 12 hourly and 500 mg 12 hourly, with a ciprofloxacin MIC of 128–256 mg/L.

PK/PD analysis

The Emax model well described the PK/PD relationship for the primary endpoint (Δlog10 cfu/mL at 72 h) with AUC0–24/MIC EI50 = 887 (R2 0.9062) and Cmax/MIC EI50 = 76 (R2 0.8766) (Figure 4). Target PK/PD ranged from 684 AUC0–24/MIC and 56 Cmax/MIC for stasis to 1521 AUC0–24/MIC and 147 Cmax/MIC for 3 log10 kill (Table 4). The PK/PD relationships for the secondary endpoints (Δlog10 cfu/mL at 96 h, and AUBKC0–96) are presented in Table 3. The best goodness of fit for data was generated using the AUBKC0–96 endpoint, with an AUC0–24/MIC EI50 = 692 (R2 0.9307) and Cmax/MIC EI50 = 57 (R2 0.9082). With MIC values measured by BMD in mSHU, the AUC0–24/MICmSHU targets for Δlog10 cfu/mL at 72 h ranged from 27 for stasis to 72 for 3 log10 kill. Similarly, the total bacterial response (AUBKC0–96) had the best goodness of fit (R2 0.9207), with the AUC0–24/MICmSHU EI50 = 30, and EI90 = 100 (Table S4 and Figure S4).

Figure 4.

Figure 4.

Exposure–response relationships. Emax models detailing the relationship between exposure [AUC0–24/MIC (left) and Cmax/MIC (right)] and the change in bacterial density, measured at (a) 72 h and (b) 96 h, and the total bacterial response (c) AUBKC0–96. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

Table 4.

Ciprofloxacin urinary PK/PD targets for different bacterial response endpoints

AUC0–24/MIC (95% CI) C max/MIC (95% CI)
End of treatment: 72 h Δlog10 cfu/mL
ȃStasis 684 (557–821) 56 (45–71)
ȃ1 log10 kill 878 (730–1063) 76 (62–100)
ȃ2 log10 kill 1125 (933–1420) 102 (80–145)
ȃ3 log10 kill 1521 (1185–2025) 147 (104–222)
ȃR2 0.9062 0.8766
Follow-up: 96 h Δlog10 cfu/mL
ȃStasis 1534 (949–2225) 123 (69–199)
ȃ1 log10 kill 2383 (1607–3306) 204 (123–313)
ȃ2 log10 kill 3651 (2584–5207) 338 (211–524)
ȃ3 log10 kill 6045 (4078–9602) 633 (374–1274)
ȃR2 0.7504 0.6927
Total bacterial response: AUBKC0–96
ȃEI50 692 (562–861) 57 (45–76)
ȃEI90 2727 (1778–4599) 253 (146–527)
ȃR2 0.9307 0.9082

Non-linear regression Emax curves described by the equation: E = (Emax − Emin) × EIn/(EIn + EIn50) + Emin, where Emax reflects maximal growth and Emin reflects no growth, EI is the exposure index of AUC0–24/MIC and Cmax/MIC, EI50 is the exposure index required to achieve 50% of Emax, EI90 is the exposure index required to achieve 90% of Emax, and n is the slope of the dose–effect relationship (Hill coefficient).

Monte Carlo simulation

PTA applying AUC0–24/MIC targets for the primary endpoint (72 h Δlog10 cfu/mL) for each oral ciprofloxacin dosing regimen is graphed overlying E. coli ciprofloxacin MIC distributions (Figure 5). A >95% PTA for a 3 log10 kill effect following 250 mg daily, 500 mg daily, 250 mg 12 hourly, 500 mg 12 hourly and 750 mg 12 hourly was found for isolates with ciprofloxacin MICs of ≤0.25, 0.5, 0.5, 1 and 1 mg/L. A >95% PTA for stasis was found for isolates with ciprofloxacin MICs of  ≤ 0.5, 1, 1, 2 and 4 mg/L for each dosing regimen, respectively (Table S5).

Figure 5.

Figure 5.

End-of-treatment (72 h) Monte Carlo simulation. PTA of stasis and log kill at 72 h for 12 hourly dosing of oral ciprofloxacin at (a) 250, (b) 500, and (c) 750 mg. Graphs overlying the ciprofloxacin MIC distribution from the 93 ceftriaxone-resistant E. coli clinical isolates (top row) and the EUCAST MIC distribution (bottom row). PTA values are shown in Table S3. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

Secondary endpoints [follow-up (96 h); total bacterial response (AUBKC0–96)] are presented in Tables S6 and S7 and Figures S5 and S6. A ciprofloxacin MIC ≤ 1 mg/L allowed for >95% PTA for a 1 log10 kill at follow-up (96 h) and 90% maximal efficacy with high (750 mg 12 hourly) dosing. For standard dosing (500 mg 12 hourly) and low dosing (250 mg 12 hourly), PK/PD breakpoints are MIC ≤ 0.5 and ≤ 0.25 mg/L for 1 log10 kill at 96 h, and MIC ≤ 0.5 and ≤ 0.25 mg/L for 90% maximal efficacy for AUBKC0–96.

Discussion

PK/PD analysis and MCS support the in vitro efficacy of high-dose ciprofloxacin (750 mg orally 12 hourly) against E. coli urinary isolates with a ciprofloxacin MIC ≤ 1 mg/L for a 3 log10 kill at the end of 3 days of treatment, 1 log10 kill at follow-up (at 96 h), and 90% maximal activity (AUBKC0–96). Standard-dose ciprofloxacin (500 mg 12 hourly) similarly achieved 3 log10 kill at 72 h at MIC ≤ 1 mg/L, whereas 1 log10 kill at 96 h and 90% maximal activity required a lower MIC of ≤0.5 mg/L. Low-dose ciprofloxacin (250 mg 12 hourly, or 500 mg daily) achieved 3 log10 kill at 72 h at MIC ≤ 0.5 mg/L, 1 log10 kill at 96 h and 90% maximal activity at MIC ≤ 0.25 mg/L. These data support current ciprofloxacin dosing practices for UTIs caused by E. coli categorized as susceptible (MIC ≤ 0.25 mg/L), and the potential to expand activity with high-dose therapy for urinary isolates with low-level ciprofloxacin resistance (MIC ≤ 1 mg/L).

Expanding ciprofloxacin susceptibility can provide an oral antimicrobial option for select groups of patients, such as: UTIs caused by MDR bacteria; oral step-down following initial IV therapy; infections following urinary tract instrumentation; infections in males; catheter-associated UTIs; and after consultation with clinical colleagues. However, given restrictions on the use of ciprofloxacin by the EMA and FDA, any change to ciprofloxacin breakpoints would need to ensure that ciprofloxacin is not promoted for uncomplicated and recurrent UTIs. Using cascade (or selective) reporting provides a valuable antimicrobial stewardship strategy for the reporting laboratory.35,36

Established ciprofloxacin target plasma PK/PD ratios are fAUC/MIC ratio of 140 for 2 log10 kill in a neutropenic thigh model and fAUC/MIC ratio of 87.5 for clinical efficacy in hospital-acquired pneumonia.37 Despite PK data recording high ciprofloxacin concentrations in urine, neither animal nor clinical PK/PD targets exist for UTIs. The UTI PK/PD targets determined in the present study were 8–16 times higher than plasma targets. This reflects a reduction in the activity of ciprofloxacin in a bladder infection model and supported by elevated MIC measurements in mSHU and pooled human urine (three to four 2-fold increase compared with testing in CAMHB). When applying ciprofloxacin MIC values measured by BMD in mSHU, the resulting urinary PK/PD targets are similar to the plasma targets in the literature. Namely, fAUC/MICmSHU ratio of 72 for 3 log10 kill at 72 h, fAUC/MICmSHU ratio of 149 for 1 log10 kill at 96 h, and fAUC/MICmSHU ratio of 100 for 90% maximal activity (AUBKC0–96). These observations support the clinical correlation of the bladder infection model to predict antibiotic efficacy in UTIs. However, applying non-standardized MIC measurements limits the practical translation of the in vitro results. The MIC should be measured by the reference method to provide a phenotypic endpoint in a defined and standardized system as a reproducible measure of antibiotic activity against that microorganism. The value should not be extrapolated as a concentration directly comparable with in vivo concentrations found at the site of infection during treatment.38

Establishing UTI-specific PK/PD targets can inform optimized antibiotic dosing strategies. However, simulating urinary PK has greater complexity compared with plasma PK given the marked variability observed between individuals, largely due to fluid intake and voiding behaviour (Table 1). Antibiotic protein binding, which is an important consideration in plasma PK, is less of an issue when considering free-drug activity and total drug measurements in urine, given the renal excretion of unbound drug and the paucity of albumin found in urine in patients with normal renal glomerular function.39,40 Urinary ciprofloxacin PK variability has been modelled in relation to urine output (1 versus 2.5 L/day), healthy young adults versus elderly, and circadian changes in diuresis and absorption.41 Such variability in urinary PK is not unique to ciprofloxacin. Wijma et al.42 found a 47% coefficient of variability (CV%) in urinary fosfomycin exposure (AUC0–48 21284 ± 9965 mg·h/L). Similarly, Wenzler et al.43 measured urinary fosfomycin concentrations at selected time periods with a CV% of 67%–84%, and a similar CV% in the volume of urine voided (60%–73%). Informed from these studies, and those presented in Table 1, we applied a ±50% variability in the expected urinary ciprofloxacin exposure (AUC0–24) in our MCS.

Mutations associated with low- and high-level ciprofloxacin resistance are complex and varied.44 In our 15 clinical E. coli isolates, ciprofloxacin MIC appeared to be related to the type and number of mutations detected. This has been similarly reported in qnr-containing E. coli with additional topoisomerase mutations and increased expression of efflux pump genes.45 Subinhibitory antibiotic concentrations have also been reported to promote resistance via target mutations and changes in drug efflux.46 An interesting observation from our data is the overall lack of emergence of ciprofloxacin resistance, determined by MIC testing at follow-up (96 h) and plating on MHA supplemented with 2 and 128 mg/L ciprofloxacin. We did not repeat WGS on the post-exposure growth or assess efflux expression. Possible explanations could be the down-regulation of genes prior to MIC testing, or that regrowth reflects tolerance, persistence and quiescence.47,48

First-line oral antimicrobials, nitrofurantoin and fosfomycin, may not be preferred agents for a variety of clinical reasons. Their activity beyond the bladder is uncertain, renal impairment impacts on nitrofurantoin,49 and fosfomycin is not reliably active against non-E. coli uropathogens.50 Alternatively, amoxicillin/clavulanate for the treatment of ESBL-producing uropathogens has been supported by several observational studies.51,52 However, a randomized control trial comparing 3 days of amoxicillin/clavulanate with 3 days of ciprofloxacin demonstrated a higher failure rate with amoxicillin/clavulanate.53 The potential superiority of fluoroquinolones over other agents, including β-lactams, has been reported in two systematic reviews.54,55

New oral antimicrobials for ESBL-producing uropathogens include: third-generation oral cephalosporins with clavulanate;56 oral carbapenems (sulopenem non-inferior to ciprofloxacin,57 tebipenem non-inferior to ertapenem58,59); omadacycline;60 gepotidacin;61 and oral β-lactamase inhibitors (QPX7728, ETX0282, VNRX7145 and ARX1796).62–64 Although these agents may provide valuable future options, the complexities of licensing new agents can limit their availability, as evidenced by an FDA regulatory hurdle for tebipenem that prompted Spero Therapeutics to suspend commercialization activities.

Our dynamic in vitro model applies a high media flow rate and large volume shifts that mimic urodynamics and mSHU to reflect the urinary environment. The addition of yeast extract and casamino acids to the 18 chemical components included in the mSHU enables the E. coli uropathogens to have a similar growth rate to growth in human urine, while not expected to bind free ciprofloxacin. However, the bactericidal activity of the ciprofloxacin is decreased by several in vivo conditions, including the presence of cations and acidic urine pH leading to higher MICs.65,66 Other dynamic PK/PD in vitro models studying ciprofloxacin against E. coli have either lacked urodynamic simulation,67 or were not specific for UTIs (i.e. urinary ciprofloxacin exposures; media mimicking urine).68,69 Where urodynamics and the urinary environment were simulated, as performed using the original UTI bladder infection in vitro model,70 and a dilutional model,71 fluoroquinolones were very effective against E. coli isolates.

Compared with in vivo studies, a mouse ascending UTI model demonstrated that isogenic E. coli strains with low-level ciprofloxacin resistance genes (qnrA1, MIC 0.19 mg/L; qnrB19, MIC 0.38 mg/L; qnrS1, MIC 0.38 mg/L) had reduced ciprofloxacin kill in urine and bladder bacterial counts compared with the WT strain (MIC 0.032 mg/L), despite ciprofloxacin reaching high urine concentrations (urinary AUC0–24 2572 mg·h/L, urinary Cmax 553 mg/L) following the dose of 0.2 mg per mouse four times daily to correspond to a human dose of 500 mg twice daily.72 In another mouse model of ascending UTI in diabetic mice, following ciprofloxacin dose-fraction studies a plasma exposure–response relationship was found with an approximate AUC/MIC IC50 of 120–170.73 A 24 h plasma AUC/MIC of ∼400 associated with complete bacterial clearance in kidneys and bladder tissues.73 Corresponding to human exposures following oral 500 mg 12 hourly, a mouse equivalent dose (196 mg/L) would be expected to provide exposures correlating with significant microbiology activity in bladder, kidney and urine and resolution of clinical symptoms (plasma AUC/MIC of 566). As urine ciprofloxacin concentrations were not measured, plasma concentrations were considered as a surrogate to evaluate therapeutic concentrations in the bladder and kidneys. This study was limited by infecting mice with the fully ciprofloxacin susceptible E. coli ATCC 25922 strain and assessing the response over 24 h.

There are important limitations to our in vitro model, including the lack of host response and bladder tissue architecture. Immunocompetent and novel ‘bladder-on-a-chip’ in vitro models have attempted to overcome some of these limitations.74,75 Our findings are also specific to E. coli and may not directly translate to other uropathogens. Furthermore, it is uncertain if these data can be extrapolated to complicated UTI, or patients with renal dysfunction. This work does not examine extended-release formulations of oral ciprofloxacin that have comparable clinical outcomes,76–78 with the benefit of daily administration, high urinary drug levels over the entire 24 h period, higher peak plasma concentrations, superior bactericidal activity and lower interpatient variability.79

In summary, these data support oral ciprofloxacin efficacy for E. coli urinary isolates with MIC ≤ 0.25 mg/L with the standard dose and with MIC ≤ 1 mg/L when dosed at 750 mg 12 hourly and applying conservative measures for bacterial response. The application of urinary-specific ciprofloxacin breakpoints should be cautiously considered in specific clinical scenarios and supported with strong antimicrobial stewardship practices.

Supplementary Material

dkac402_Supplementary_Data

Acknowledgments

We thank Dr Adam Jenney, Microbiology Unit, Department of Pathology, Alfred Health, Melbourne, Australia for providing the baseline collection of E. coli clinical isolates.

Contributor Information

Iain J Abbott, Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia.

Elke van Gorp, Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia.

Hugh Cottingham, Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia.

Nenad Macesic, Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia.

Steven C Wallis, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia.

Jason A Roberts, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia; Department of Intensive Care Medicine and Pharmacy Department, Royal Brisbane and Women’s Hospital, Brisbane, Australia; Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France.

Joseph Meletiadis, Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Haidari, Athens, Greece.

Anton Y Peleg, Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia; Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia.

Funding

This study was supported by internal funding. A.Y.P. is part funded through an Australian National Health and Medical Research Council (NHMRC) Practitioner Fellowships (APP1117065). J.A.R. would like to acknowledge funding from the Australian National Health and Medical Research Council for a Centre of Research Excellence (APP2007007) and an Investigator Grant (APP2009736) as well as an Advancing Queensland Clinical Fellowship.

Transparency declarations

I.J.A. has received consultancies/advisory boards for MSD. J.M. has received research grants from Pfizer, Gilead, Astellas, MSD, F2G and Venatorx. J.A.R. has consultancies/advisory boards for MSD, QPEX, Discuva Ltd, Accelerate Diagnostics, Bayer, bioMérieux; speaking fees for MSD, bioMérieux, Pfizer; industry grants from MSD, The Medicines Company, Cardeas Pharma, bioMérieux, QPEX, Pfizer. A.Y.P. has received investigator-initiated research funding from MSD. All other authors: none to declare.

Supplementary data

Supplementary Methods, Figures S1 and S6 and Tables S1 to S7 are available as Supplementary data at JAC Online.

References

  • 1. Flores-Mireles AL, Walker JN, Caparon Met al. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat Rev Microbiol 2015; 13: 269–84. 10.1038/nrmicro3432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Keijzers G, Macdonald SP, Udy AAet al. The Australasian resuscitation in sepsis evaluation: fluids or vasopressors in emergency department sepsis (ARISE FLUIDS), a multi-centre observational study describing current practice in Australia and New Zealand. Emerg Med Australas 2020; 32: 586–98. 10.1111/1742-6723.13469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Jauneikaite E, Honeyford K, Blandy Oet al. Bacterial genotypic and patient risk factors for adverse outcomes in Escherichia coli bloodstream infections: a prospective molecular epidemiological study. J Antimicrob Chemother 2022; 77: 1753–61. 10.1093/jac/dkac071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Pitout JDD, Peirano G, Chen Let al. Escherichia coli ST1193: following in the footsteps of E. coli ST131. Antimicrob Agents Chemother 2022; 66: e0051122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Raja NS. Oral treatment options for patients with urinary tract infections caused by extended spectrum βeta-lactamase (ESBL) producing Enterobacteriaceae. J Infect Public Health 2019; 12: 843–6. 10.1016/j.jiph.2019.05.012 [DOI] [PubMed] [Google Scholar]
  • 6. CDC . Antibiotic Resistance Threats in the United States, 2019. 2019. https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threats-report-508.pdf. [Google Scholar]
  • 7. Tutone M, Bjerklund Johansen TE, Cai Tet al. Susceptibility and resistance to fosfomycin and other antimicrobial agents among pathogens causing lower urinary tract infections: findings of the SURF study. Int J Antimicrob Agents 2022; 59: 106574. 10.1016/j.ijantimicag.2022.106574 [DOI] [PubMed] [Google Scholar]
  • 8. Cassini A, Hogberg LD, Plachouras Det al. Attributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the European Economic Area in 2015: a population-level modelling analysis. Lancet Infect Dis 2019; 19: 56–66. 10.1016/S1473-3099(18)30605-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Bezabih YM, Sabiiti W, Alamneh Eet al. The global prevalence and trend of human intestinal carriage of ESBL-producing Escherichia coli in the community. J Antimicrob Chemother 2021; 76: 22–9. 10.1093/jac/dkaa399 [DOI] [PubMed] [Google Scholar]
  • 10. Aronin SI, Gupta V, Dunne MWet al. Regional differences in antibiotic-resistant Enterobacterales urine isolates in the United States: 2018-2020. Int J Infect Dis 2022; 119: 142–5. 10.1016/j.ijid.2022.03.052 [DOI] [PubMed] [Google Scholar]
  • 11. Antimicrobial Resistance Collaborators . Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 2022; 399: 629–55. 10.1016/S0140-6736(21)02724-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Li X, Fan H, Zi Het al. Global and regional burden of bacterial antimicrobial resistance in urinary tract infections in 2019. J Clin Med 2022; 11: 2817. 10.3390/jcm11102817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Aronoff GE, Kenner CH, Sloan RSet al. Multiple-dose ciprofloxacin kinetics in normal subjects. Clin Pharmacol Ther 1984; 36: 384–8. 10.1038/clpt.1984.192 [DOI] [PubMed] [Google Scholar]
  • 14. Gonzalez MA, Uribe F, Moisen SDet al. Multiple-dose pharmacokinetics and safety of ciprofloxacin in normal volunteers. Antimicrob Agents Chemother 1984; 26: 741–4. 10.1128/AAC.26.5.741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Hoffken G, Lode H, Prinzing Cet al. Pharmacokinetics of ciprofloxacin after oral and parenteral administration. Antimicrob Agents Chemother 1985; 27: 375–9. 10.1128/AAC.27.3.375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Joos B, Ledergerber B, Flepp Met al. Comparison of high-pressure liquid chromatography and bioassay for determination of ciprofloxacin in serum and urine. Antimicrob Agents Chemother 1985; 27: 353–6. 10.1128/AAC.27.3.353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Davis RL, Koup JR, Williams-Warren Jet al. Pharmacokinetics of three oral formulations of ciprofloxacin. Antimicrob Agents Chemother 1985; 28: 74–7. 10.1128/AAC.28.1.74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Tartaglione TA, Raffalovich AC, Poynor WJet al. Pharmacokinetics and tolerance of ciprofloxacin after sequential increasing oral doses. Antimicrob Agents Chemother 1986; 29: 62–6. 10.1128/AAC.29.1.62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Catchpole C, Andrews JM, Woodcock Jet al. The comparative pharmacokinetics and tissue penetration of single-dose ciprofloxacin 400 mg iv and 750 mg po. J Antimicrob Chemother 1994; 33: 103–10. 10.1093/jac/33.1.103 [DOI] [PubMed] [Google Scholar]
  • 20. Wagenlehner FM, Wydra S, Onda Het al. Concentrations in plasma, urinary excretion, and bactericidal activity of linezolid (600 milligrams) versus those of ciprofloxacin (500 milligrams) in healthy volunteers receiving a single oral dose. Antimicrob Agents Chemother 2003; 47: 3789–94. 10.1128/AAC.47.12.3789-3794.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Boy D, Well M, Kinzig-Schippers Met al. Urinary bactericidal activity, urinary excretion and plasma concentrations of gatifloxacin (400 mg) versus ciprofloxacin (500 mg) in healthy volunteers after a single oral dose. Int J Antimicrob Agents 2004; 23Suppl 1: S6–16. 10.1016/j.ijantimicag.2003.09.006 [DOI] [PubMed] [Google Scholar]
  • 22. Wagenlehner FM, Kinzig-Schippers M, Sorgel Fet al. Concentrations in plasma, urinary excretion and bactericidal activity of levofloxacin (500 mg) versus ciprofloxacin (500 mg) in healthy volunteers receiving a single oral dose. Int J Antimicrob Agents 2006; 28: 551–9. 10.1016/j.ijantimicag.2006.07.026 [DOI] [PubMed] [Google Scholar]
  • 23. US FDA . Drug Safety Communication: FDA warns about increased risk of ruptures or tears in the aorta blood vessel with fluoroquinolone antibiotics in certain patients. 20 December 2018. https://www.fda.gov/Drugs/DrugSafety/ucm628753.htm.
  • 24. EMA . Disabling and potentially permanent side effects lead to suspension or restrictions of quinolone and fluoroquinolone antibiotics. 11 March 2019. https://www.ema.europa.eu/en/medicines/human/referrals/quinolone-fluoroquinolone-containing-medicinal-products.
  • 25. EUCAST . Breakpoint tables for interpretation of MICs and zone diameters. Version 12.0, 2022. http://www.eucast.org.
  • 26. CLSI . Performance Standards for Antimicrobial Susceptibility Testing—Thirty-Second Edition: M100. 2022. [Google Scholar]
  • 27. Abbott IJ, Peel TN, Cairns KAet al. Antibiotic management of urinary tract infections in the post-antibiotic era: a narrative review highlighting diagnostic and antimicrobial stewardship. Clin Microbiol Infect 2022: S1198–743X(22)00272–5. 10.1016/j.cmi.2022.05.016 [DOI] [PubMed] [Google Scholar]
  • 28. Stapleton AE, Wagenlehner FME, Mulgirigama Aet al. Escherichia coli resistance to fluoroquinolones in community-acquired uncomplicated urinary tract infection in women: a systematic review. Antimicrob Agents Chemother 2020; 64: e00862-20. 10.1128/AAC.00862-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Ipe DS, Ulett GC. Evaluation of the in vitro growth of urinary tract infection-causing gram-negative and gram-positive bacteria in a proposed synthetic human urine (SHU) medium. J Microbiol Methods 2016; 127: 164–71. 10.1016/j.mimet.2016.06.013 [DOI] [PubMed] [Google Scholar]
  • 30. ISO . ISO 20776-1:2006. Clinical Laboratory Testing and In Vitro Diagnostic Test Systems - Susceptibility Testing of Infectious Agents and Evaluation of Performance of Antimicrobial Susceptibility Testing Devices- Part 1. https://www.iso.org/standard/41630.html. [Google Scholar]
  • 31. Abbott IJ, van Gorp E, Wyres KLet al. Oral fosfomycin activity against Klebsiella pneumoniae in a dynamic bladder infection in vitro model. J Antimicrob Chemother 2022; 77: 1324–33. 10.1093/jac/dkac045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Rowe EL, Morozowich W. A simple dilution analog computer for simulation of drug distribution processes. J Pharm Sci 1969; 58: 1375–8. 10.1002/jps.2600581117 [DOI] [PubMed] [Google Scholar]
  • 33. FDA . Bioanalytical Method Validation: Guidance for Industry. 2018. https://www.fda.gov/media/70858/download.
  • 34. EUCAST . EUCAST MIC distribution website. https://mic.eucast.org.
  • 35. Kahlmeter G, Thilly N, Pulcini C. Selective reporting of antibiotic susceptibility testing results: less is more. Clin Microbiol Infect 2021; 27: 503–5. 10.1016/j.cmi.2020.11.017 [DOI] [PubMed] [Google Scholar]
  • 36. Tebano G, Mouelhi Y, Zanichelli Vet al. Selective reporting of antibiotic susceptibility testing results: a promising antibiotic stewardship tool. Expert Rev Anti Infect Ther 2020; 18: 251–62. 10.1080/14787210.2020.1715795 [DOI] [PubMed] [Google Scholar]
  • 37. EUCAST . Ciprofloxacin Rationale Document, version 2.0, 2021. https://www.eucast.org/publications_and_documents/rd/.
  • 38. Mouton JW, Muller AE, Canton Ret al. MIC-based dose adjustment: facts and fables. J Antimicrob Chemother 2018; 73: 564–8. 10.1093/jac/dkx427 [DOI] [PubMed] [Google Scholar]
  • 39. Ahmed H, Bergmann F, Zeitlinger M. Protein binding in translational antimicrobial development-focus on interspecies differences. Antibiotics (Basel) 2022; 11: 923. 10.3390/antibiotics11070923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zeitlinger MA, Derendorf H, Mouton JWet al. Protein binding: do we ever learn? Antimicrob Agents Chemother 2011; 55: 3067–74. 10.1128/AAC.01433-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Sanchez Navarro MD, Sayalero Marinero ML, Sanchez Navarro A. Pharmacokinetic/pharmacodynamic modelling of ciprofloxacin 250 mg/12 h versus 500 mg/24 h for urinary infections. J Antimicrob Chemother 2002; 50: 67–72. 10.1093/jac/dkf079 [DOI] [PubMed] [Google Scholar]
  • 42. Wijma RA, Koch BCP, van Gelder Tet al. High interindividual variability in urinary fosfomycin concentrations in healthy female volunteers. Clin Microbiol Infect 2018; 24: 528–32. 10.1016/j.cmi.2017.08.023 [DOI] [PubMed] [Google Scholar]
  • 43. Wenzler E, Ellis-Grosse EJ, Rodvold KA. Pharmacokinetics, safety, and tolerability of single-dose intravenous (ZTI-01) and oral fosfomycin in healthy volunteers. Antimicrob Agents Chemother 2017; 61: e00775-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Morgan-Linnell SK, Becnel Boyd L, Steffen Det al. Mechanisms accounting for fluoroquinolone resistance in Escherichia coli clinical isolates. Antimicrob Agents Chemother 2009; 53: 235–41. 10.1128/AAC.00665-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Vinue L, Hooper DC, Jacoby GA. Chromosomal mutations that accompany qnr in clinical isolates of Escherichia coli. Int J Antimicrob Agents 2018; 51: 479–83. 10.1016/j.ijantimicag.2018.01.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Ching C, Orubu ESF, Sutradhar Iet al. Bacterial antibiotic resistance development and mutagenesis following exposure to subinhibitory concentrations of fluoroquinolones in vitro: a systematic review of the literature. JAC Antimicrob Resist 2020; 2: dlaa068. 10.1093/jacamr/dlaa068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Ortiz-Padilla M, Diaz-Diaz S, Machuca Jet al. Role of low-level quinolone resistance in generating tolerance in Escherichia coli under therapeutic concentrations of ciprofloxacin. J Antimicrob Chemother 2020; 75: 2124–32. 10.1093/jac/dkaa151 [DOI] [PubMed] [Google Scholar]
  • 48. Leatham-Jensen MP, Mokszycki ME, Rowley DCet al. Uropathogenic Escherichia coli metabolite-dependent quiescence and persistence may explain antibiotic tolerance during urinary tract infection. mSphere 2016; 1: e00055-15. 10.1128/mSphere.00055-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Ten Doesschate T, van Haren E, Wijma RAet al. The effectiveness of nitrofurantoin, fosfomycin and trimethoprim for the treatment of cystitis in relation to renal function. Clin Microbiol Infect 2020; 26: 1355–60. 10.1016/j.cmi.2020.03.001 [DOI] [PubMed] [Google Scholar]
  • 50. Abbott IJ, Mouton JW, Peleg AYet al. Pharmacokinetic/pharmacodynamic analysis of oral fosfomycin against Enterobacterales, Pseudomonas aeruginosa and Enterococcus spp. in an in vitro bladder infection model: impact on clinical breakpoints. J Antimicrob Chemother 2021; 76: 3201–11. 10.1093/jac/dkab313 [DOI] [PubMed] [Google Scholar]
  • 51. Beytur A, Yakupogullari Y, Oguz Fet al. Oral amoxicillin-clavulanic acid treatment in urinary tract infections caused by extended-spectrum beta-lactamase-producing organisms. Jundishapur J Microbiol 2015; 8: e13792. 10.5812/jjm.13792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Rodriguez-Bano J, Navarro MD, Retamar Pet al. β-Lactam/β-lactam inhibitor combinations for the treatment of bacteremia due to extended-spectrum β-lactamase-producing Escherichia coli: a post hoc analysis of prospective cohorts. Clin Infect Dis 2012; 54: 167–74. 10.1093/cid/cir790 [DOI] [PubMed] [Google Scholar]
  • 53. Hooton TM, Scholes D, Gupta Ket al. Amoxicillin-clavulanate vs ciprofloxacin for the treatment of uncomplicated cystitis in women: a randomized trial. JAMA 2005; 293: 949–55. 10.1001/jama.293.8.949 [DOI] [PubMed] [Google Scholar]
  • 54. Punjabi C, Tien V, Meng Let al. Oral fluoroquinolone or trimethoprim-sulfamethoxazole vs. β-lactams as step-down therapy for Enterobacteriaceae bacteremia: systematic review and meta-analysis. Open Forum Infect Dis 2019; 6: ofz364. 10.1093/ofid/ofz364 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Yan K, Zhu M, Jia Yet al. Efficacy and safety of quinolones vs. other antimicrobials for the treatment of uncomplicated urinary tract infections in adults: a systematic review and meta-analysis. Int Urogynecol J 2022; 33: 1103–23. 10.1007/s00192-021-05013-4 [DOI] [PubMed] [Google Scholar]
  • 56. Stewart AG, Harris PNA, Henderson Aet al. Oral cephalosporin and β-lactamase inhibitor combinations for ESBL-producing Enterobacteriaceae urinary tract infections. J Antimicrob Chemother 2020; 75: 2384–93. 10.1093/jac/dkaa183 [DOI] [PubMed] [Google Scholar]
  • 57. Dunne MW, Das AF, Zelasky Met al. LB-1. Efficacy and safety of oral sulopenem etzadroxil/probenecid versus oral ciprofloxacin in the treatment of uncomplicated urinary tract infections (uUTI) in adult women: results from the SURE-1 trial. Open Forum Infect Dis 2020; 7: S844-S. 10.1093/ofid/ofaa515.1898 [DOI] [Google Scholar]
  • 58. Muir LA, Walpole SM, Warfel PAet al. LB-3. Oral tebipenem pivoxil hydrobromide is non-inferior to IV ertapenem in complicated urinary tract infection (cUTI) and acute pyelonephritis (AP) – results from the pivotal ADAPT-PO study. Open Forum Infect Dis 2020; 7: S844–S5. 10.1093/ofid/ofaa515.1900 [DOI] [Google Scholar]
  • 59. Eckburg PB, Muir L, Critchley IAet al. Oral tebipenem pivoxil hydrobromide in complicated urinary tract infection. N Engl J Med 2022; 386: 1327–38. 10.1056/NEJMoa2105462 [DOI] [PubMed] [Google Scholar]
  • 60. Overcash JS, Bhiwandi P, Garrity-Ryan Let al. Pharmacokinetics, safety, and clinical outcomes of omadacycline in women with cystitis: results from a phase 1b study. Antimicrob Agents Chemother 2019; 63: e02083-18. 10.1128/AAC.02083-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Scangarella-Oman NE, Hossain M, Hoover JLet al. Dose selection for phase III clinical evaluation of gepotidacin (GSK2140944) in the treatment of uncomplicated urinary tract infections. Antimicrob Agents Chemother 2022; 66: e0149221. 10.1128/aac.01492-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Chatwin CL, Hamrick JC, Trout RELet al. Microbiological characterization of VNRX-5236, a broad-spectrum β-lactamase inhibitor for rescue of the orally bioavailable cephalosporin ceftibuten as a carbapenem-sparing agent against strains of Enterobacterales expressing extended-spectrum β-lactamases and serine carbapenemases. Antimicrob Agents Chemother 2021; 65: e0055221. 10.1128/AAC.00552-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Veeraraghavan B, Bakthavatchalam YD, Sahni RD. Oral antibiotics in clinical development for community-acquired urinary tract infections. Infect Dis Ther 2021; 10: 1815–35. 10.1007/s40121-021-00509-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Lomovskaya O, Rubio-Aparicio D, Tsivkovski Ret al. The ultra-broad-spectrum beta-lactamase inhibitor QPX7728 restores the potency of multiple oral beta-lactam antibiotics against beta-lactamase producing strains of resistant Enterobacterales. Antimicrob Agents Chemother 2022; 66: e0216821. 10.1128/AAC.02168-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Smith JT, Ratcliffe NT. Effect of pH value and magnesium on the antibacterial activity of quinolone preparations. Infection 1986; 14Suppl 1: S31–5. 10.1007/BF01645195 [DOI] [PubMed] [Google Scholar]
  • 66. Kamberi M, Tsutsumi K, Kotegawa Tet al. Influences of urinary pH on ciprofloxacin pharmacokinetics in humans and antimicrobial activity in vitro versus those of sparfloxacin. Antimicrob Agents Chemother 1999; 43: 525–9. 10.1128/AAC.43.3.525 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Drobot GR, Karlowsky JA, Hoban DJet al. Antibiotic activity in microbiological media versus that in human urine: comparison of ampicillin, ciprofloxacin, and trimethoprim-sulfamethoxazole. Antimicrob Agents Chemother 1996; 40: 237–40. 10.1128/AAC.40.1.237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Firsov AA, Zinner SH, Lubenko Iet al. Gemifloxacin and ciprofloxacin pharmacodynamics in an in-vitro dynamic model: prediction of the equivalent AUC/MIC breakpoints and doses. Int J Antimicrob Agents 2000; 16: 407–14. 10.1016/S0924-8579(00)00226-0 [DOI] [PubMed] [Google Scholar]
  • 69. Vostrov SN, Kononenko OV, Lubenko IYet al. Comparative pharmacodynamics of gatifloxacin and ciprofloxacin in an in vitro dynamic model: prediction of equiefficient doses and the breakpoints of the area under the curve/MIC ratio. Antimicrob Agents Chemother 2000; 44: 879–84. 10.1128/AAC.44.4.879-884.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Greenwood D, Baxter S, Cowlishaw Aet al. Antibacterial activity of ciprofloxacin in conventional tests and in a model of bacterial cystitis. Eur J Clin Microbiol 1984; 3: 351–4. 10.1007/BF01977493 [DOI] [PubMed] [Google Scholar]
  • 71. Nishimura M, Kumamoto Y, Shibuya Aet al. An in vitro study on the treatment of complicated cystitis using an automatic simulator. Kansenshogaku Zasshi 1990; 64: 1004–12. 10.11150/kansenshogakuzasshi1970.64.1004 [DOI] [PubMed] [Google Scholar]
  • 72. Jakobsen L, Cattoir V, Jensen KSet al. Impact of low-level fluoroquinolone resistance genes qnrA1, qnrB19 and qnrS1 on ciprofloxacin treatment of isogenic Escherichia coli strains in a murine urinary tract infection model. J Antimicrob Chemother 2012; 67: 2438–44. 10.1093/jac/dks224 [DOI] [PubMed] [Google Scholar]
  • 73. Reniguntla MK, Yedle R, Puttaswamy Ret al. Pharmacokinetics/pharmacodynamics (PK/PD) of ciprofloxacin in the complicated urinary tract infection (cUTI) model in diabetic mice. Curr Drug Metab 2020; 21: 132–9. 10.2174/1389200221666200310105227 [DOI] [PubMed] [Google Scholar]
  • 74. Murray BO, Flores C, Williams Cet al. Recurrent urinary tract infection: a mystery in search of better model systems. Front Cell Infect Microbiol 2021; 11: 691210. 10.3389/fcimb.2021.691210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Dalhoff A. Contribution of immunocompetence to the antibacterial activities of ciprofloxacin and moxifloxacin in an in vitro pharmacodynamic model. Infection 2005; 33Suppl 2: 44–9. 10.1007/s15010-005-8207-x [DOI] [PubMed] [Google Scholar]
  • 76. Henry DC Jr, Bettis RB, Riffer Eet al. Comparison of once-daily extended-release ciprofloxacin and conventional twice-daily ciprofloxacin for the treatment of uncomplicated urinary tract infection in women. Clin Ther 2002; 24: 2088–104. 10.1016/S0149-2918(02)80099-6 [DOI] [PubMed] [Google Scholar]
  • 77. Fourcroy JL, Berner B, Chiang YKet al. Efficacy and safety of a novel once-daily extended-release ciprofloxacin tablet formulation for treatment of uncomplicated urinary tract infection in women. Antimicrob Agents Chemother 2005; 49: 4137–43. 10.1128/AAC.49.10.4137-4143.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Talan DA, Klimberg IW, Nicolle LEet al. Once daily, extended release ciprofloxacin for complicated urinary tract infections and acute uncomplicated pyelonephritis. J Urol 2004; 171: 734–9. 10.1097/01.ju.0000106191.11936.64 [DOI] [PubMed] [Google Scholar]
  • 79. Talan DA, Naber KG, Palou Jet al. Extended-release ciprofloxacin (Cipro XR) for treatment of urinary tract infections. Int J Antimicrob Agents 2004; 23Suppl 1: S54–66. 10.1016/j.ijantimicag.2003.12.005 [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

dkac402_Supplementary_Data

Articles from Journal of Antimicrobial Chemotherapy are provided here courtesy of Oxford University Press

RESOURCES