ABSTRACT
SPR206 is a next-generation polymyxin being developed for the treatment of multidrug-resistant (MDR) Gram-negative infections. This Phase 1 bronchoalveolar lavage (BAL) study was conducted to evaluate SPR206’s safety and pharmacokinetics in plasma, pulmonary epithelial lining fluid (ELF), and alveolar macrophages (AM) in healthy volunteers. Subjects received a 100 mg intravenous (IV) dose of SPR206 infused over 1 h every 8 h for 3 consecutive doses. Each subject underwent 1 bronchoscopy with BAL at 2, 3, 4, 6, or 8 h after the start of the third IV infusion. SPR206 concentrations in plasma, BAL, and cell pellet were measured with a validated LC-MS/MS assay. Thirty-four subjects completed the study and 30 completed bronchoscopies. Mean SPR206 peak concentrations (Cmax) in plasma, ELF, and AM were 4395.0, 735.5, and 860.6 ng/mL, respectively. Mean area under the concentration-time curve (AUC0-8) for SPR206 in plasma, ELF, and AM was 20120.7, 4859.8, and 6026.4 ng*h/mL, respectively. The mean ELF to unbound plasma concentration ratio was 0.264, and mean AM to unbound plasma concentration ratio was 0.328. Mean SPR206 concentrations in ELF achieved lung exposures above the MIC for target Gram-negative pathogens for the entire 8-h dosing interval. Overall, SPR206 was well tolerated; 22 subjects (64.7%) reported at least 1 treatment-emergent adverse event (TEAE). Of the 40 reported TEAEs, 34 (85.0%) were reported as mild in severity. The most frequent TEAEs were oral paresthesia (10 subjects [29.4%]) and nausea (2 subjects [5.9%]). This study demonstrates pulmonary penetration of SPR206 and supports further development of SPR206 for the treatment of patients with serious infections caused by MDR Gram-negative pathogens.
KEYWORDS: SPR206, alveolar macrophage, epithelial lining fluid, pharmacokinetics, pulmonary
INTRODUCTION
Antimicrobial resistance is a growing problem worldwide, and carbapenem-resistant Acinetobacter baumannii, multidrug-resistant (MDR) Pseudomonas aeruginosa, and carbapenem-resistant and extended-spectrum beta-lactamase (ESBL)-producing Enterobacterales have been identified as urgent or serious threats by the CDC and WHO (1, 2). In particular, bacteremia, hospital-acquired and ventilator-associated bacterial pneumonia (HABP/VABP), and complicated urinary tract infections are often caused by A. baumannii (3), frequently MDR strains (4–7), and these infections account for excess morbidity and mortality (3, 8–15). Similarly, P. aeruginosa causes serious systemic infections with increased rates of morbidity and mortality (16, 17).
The continuing emergence and spread of MDR ESBL- and carbapenemase-producing clinical isolates of Enterobacterales, P. aeruginosa, and A. baumannii are limiting available options to treat infections caused by these pathogens. At present, most carbapenemase-producing strains of Klebsiella pneumoniae, Escherichia coli, P. aeruginosa, and A. baumannii remain susceptible to polymyxin antibiotics (polymyxin B and colistin), and a resurgence is seen in the use of these drugs for the treatment of serious Gram-negative infections. However, toxicity, particularly nephrotoxicity and neurotoxicity, restricts the use of polymyxins (18, 19). These findings highlight the urgent need to identify new antimicrobial agents to treat serious infections due to MDR Gram-negative pathogens (1, 2, 20–24).
SPR206 is a polymyxin derivative with activity against many Gram-negative pathogens, including A. baumannii, P. aeruginosa, K. pneumoniae, E. coli, and Enterobacter spp. that pose an increased risk for antimicrobial resistance (25–30). In vitro and in vivo studies suggest that the activity of SPR206 is more potent compared to polymyxin B (31–33). Additionally, nonclinical studies suggest the potential for an improved safety profile with SPR206 compared to polymyxin B (25, 34, 35). SPR206 was generally safe and well tolerated at exposures required for efficacy, with a low risk for respiratory, central nervous system, or cardiovascular events and a low risk for clinically significant drug-drug interactions (34, 35). SPR206 is being developed for intravenous (IV) administration for the treatment of serious infections of the respiratory tract, namely, HABP/VABP due to drug-resistant Gram-negative pathogens. A first-in-human pharmacokinetic (PK) and safety study demonstrated no appreciable drug accumulation with repeated 100 mg IV doses q8h of SPR206 for 14 days and no evidence of nephrotoxicity (36).
Concentrations of antibiotics in pulmonary epithelial lining fluid (ELF) and in alveolar macrophages (AM) are important for determining antibiotic activity and optimal dosing in patients with pneumonia (37, 38). This study was designed to determine the concentrations of SPR206 in ELF and AM compartments of the lung to provide essential information for the development of SPR206 as an antibacterial agent for the treatment of lower respiratory tract infections. The primary objectives of this study were to evaluate the safety and the intrapulmonary PK, including ELF and AM concentrations, of SPR206 compared to plasma concentrations of SPR206 in healthy adult subjects.
RESULTS
Thirty-four subjects were enrolled in the study, and all were dosed and included in the safety and PK populations. Seven subjects were excluded from the BAL PK analysis for protocol deviations (3) or lack of completed BAL samples (4), leaving 27 subjects in the BAL PK analysis. Baseline characteristics are shown in Table 1.
TABLE 1.
Baseline characteristics (safety population)
| Characteristic | Subjects (N = 34) |
|---|---|
| Age, yrsa | 41.9 ± 8.7 |
| Age range, yrs | 24–55 |
| Male, n (%) | 24 (70.6) |
| Race, n (%) | |
| Asian | 3 (8.8) |
| White | 30 (88.2) |
| Not reported | 1 (2.9) |
| Not Hispanic or Latino, n (%) | 32 (94.1) |
| wt, kga | 79.4 ± 12.4 |
| Body mass index, kg/m2a | 26.4 ± 2.8 |
Mean ± standard deviation.
Pharmacokinetics.
Following administration of SPR206 100 mg IV q8h, plasma SPR206 concentrations peaked at 2 h following the third dose, and thereafter declined reaching pre-dose concentrations at 8 h after the third dose (Fig. 1). The similarity of SPR206 concentrations prior to the third dose and at 8 h following that dose indicated that steady-state had been achieved. Median time to peak plasma concentrations (Tmax) occurred at 2 h (range: 1.8 h to 2.9 h) (Table 2).
FIG 1.
Mean (SD) plasma (total) SPR206 concentrations after 100 mg intravenously every 8 h for 3 doses.
TABLE 2.
Plasma pharmacokinetic (PK) parameters obtained after the third SPR206 100 mg intravenous dose (PK population)a
| Parameter | Arithmetic mean (SDb) | Geometric mean (CVc%) |
|---|---|---|
| AUC0-8 (h*ng/mL)d | 20120.7 (3555.2) | 19817.9 (17.9) |
| Cmaxe (ng/mL) | 4395.0 (752.7) | 4333.4 (17.2) |
| Cminf (ng/mL) | 1409.9 (709.4) | 1306.9 (36.7) |
| Tmaxg (hours) | 2.1 (0.2) | 2.1 (7.4) |
| t1/2h (hours) | 3.6 (0.5) | 3.6 (13.5) |
| Vzi (mL) | 19806.1 (3533.7) | 19502.1 (18.1) |
PK population included all the subjects who received at least one dose of SPR206 and had at least one evaluable plasma concentration. d, h, and i, N = 32; e, f, and g, N = 34.
SD, standard deviation.
CV, coefficient of variation.
AUC0-8, area under the concentration-time curve from time zero to 8 h.
Cmax, maximum plasma concentration.
Cmin, minimum plasma concentration.
Tmax, time to Cmax.
t1/2, half-life.
Vz, volume of distribution at the terminal phase.
In the BAL PK Population, 3 doses of SPR206 100 mg IV q8h produced steady-state concentrations in ELF and AM (Fig. 2) with concentration-time profiles of SPR206 for ELF and AM that were nearly flat. At each post-dose BAL sampling time point, the mean SPR206 concentration in AM (range: 604.2 ng/mL to 860.6 ng/mL) was slightly higher than the ELF concentration (range: 431.5 ng/mL to 735.5 ng/mL) (Table 3). Mean area under the concentration-time curve from time zero to 8 h (AUC0-8) for SPR206 in ELF and AM was 4859.8 h*ng/mL and 6026.4 h*ng/mL, respectively. Mean peak concentrations (Cmax) for SPR206 in ELF and AM were 735.5 ng/mL and 860.6 ng/mL, respectively, and mean minimum concentration (Cmin) in ELF and AM was 431.5 ng/mL and 604.2 ng/mL, respectively. The Tmax in ELF was 2.2 h; however, the Tmax was delayed to 6.4 h in AM.
FIG 2.
Mean (SD) SPR206 concentrations in plasma (total and unbound), ELF, and AM at BAL sampling time points after the third 100 mg intravenous dose. AM, alveolar macrophages; ELF, epithelial lining fluid.
TABLE 3.
Ratio of ELF and AM concentrations to total and unbound plasma concentrations of SPR206a
| Arithmetic mean ± SD |
||||
|---|---|---|---|---|
| BALb sampling time post-dose (hours) | ELFc to total plasma | ELF to unbound plasma | AMd to total plasma | AM to unbound plasma |
| 2 | 0.183 ± 0.032 | 0.200 ± 0.035 | 0.206 ± 0.114 | 0.226 ± 0.124 |
| 3 | 0.199 ± 0.066 | 0.218 ± 0.072 | 0.184 ± 0.065 | 0.201 ± 0.071 |
| 4 | 0.190 ± 0.067 | 0.208 ± 0.074 | 0.223 ± 0.089 | 0.244 ± 0.097 |
| 6 | 0.428 ± 0.176 | 0.469 ± 0.192 | 0.519 ± 0.223 | 0.568 ± 0.244 |
| 8 | 0.347 ± 0.138 | 0.380 ± 0.151 | 0.503 ± 0.192 | 0.550 ± 0.210 |
PK Population included all the subjects who received at least one dose of SPR206 and had at least one evaluable plasma concentration. The BAL PK Population included all the subjects who received at least one dose of SPR206 and had at least one evaluable BAL concentration. Ratios were calculated by dividing individual SPR206 concentration in ELF or AM by corresponding total and unbound plasma SPR206 concentration.
BAL, bronchoalveolar lavage.
ELF, epithelial lining fluid.
AM, alveolar macrophages.
Both ELF and AM concentrations were lower than total and unbound SPR206 plasma concentrations over the 8-h sampling period after the last dose (Fig. 2). SPR206 penetration ratios in ELF and AM relative to unbound plasma concentrations ranged from 0.200 to 0.469 for ELF and 0.201 to 0.568 for AM (Table 3). Furthermore, the ratios of SPR206 in AM to unbound plasma concentrations were slightly higher compared with ratios of ELF to unbound plasma concentrations for most BAL sampling time points (Table 4).
TABLE 4.
Ratios of ELF and AM AUC0-8 to total and unbound plasma AUC0-8 following SPR206a
| Ratio of AUC0-8b of ELFc to AUC0-8 of total plasma | Ratio of AUC0-8 of ELF to AUC0-8 of unbound plasma | Ratio of AUC0-8 of AMd to AUC0-8 of total plasma | Ratio of AUC0-8 of AM to AUC0-8 of unbound plasma |
|---|---|---|---|
| 0.242 | 0.264 | 0.300 | 0.328 |
PK Population included all the subjects who received at least one dose of SPR206 and had at least one evaluable plasma concentration. The BALe PK Population included all the subjects who received at least one dose of SPR206 and had at least one evaluable BAL concentration. The AUC0-8 of total and unbound plasma was calculated using PK Population (n = 34). The AUC0-8 of ELF and AM was calculated using BAL PK Population (n = 27).
AUC0-8, area under the concentration-time curve from time zero to 8 h.
ELF, epithelial lining fluid.
AM, alveolar macrophages.
BAL, bronchoalveolar lavage.
Safety.
Treatment with SPR206 was safe and well tolerated in healthy subjects. Overall, 64.7% of subjects experienced at least 1 treatment-emergent adverse event (TEAE) and 13 subjects (38.2%) experienced at least 1 treatment-related TEAE. Of the 40 reported TEAEs, 34 (85.0%) were reported as mild in severity. The most frequent treatment-related TEAEs were oral paresthesia in 10 (29.4%) subjects and nausea in 2 (5.9%) subjects. All paresthesia-like events (paresthesia or hypoesthesia) were mild. No deaths, serious AEs, or AEs leading to study drug discontinuation or study discontinuation were reported. No clinically significant abnormalities were reported for clinical laboratory parameters including blood urea nitrogen, serum creatinine, and estimated creatinine clearance. No clinically significant changes were noted for vital signs, electrocardiogram (ECG) assessments, or physical examinations.
DISCUSSION
The PK analysis of SPR206 plasma concentrations in this study are consistent with previous findings from a Phase 1 ascending dose study of SPR206 in healthy subjects (36). In the single SPR206 100 mg IV dose cohort, Cmax was 5300 ng/mL and AUC0-inf was 20400 h*ng/mL, which was comparable to the Cmax of 4300 ng/mL and steady-state AUC0-8 of 19800 h*ng/mL reported here. The lower values of Cmax and AUC0-8 observed in this study were likely explained by differences in the number blood samples (13 versus 6) and the absence of a sampling time at the end of the 1-h infusion. The data from this study will be incorporated into a population PK model under development and assessed relative to pharmacokinetic/pharmacodynamic targets for efficacy to provide additional support for probability of PK target attainment and dose selection for future studies.
The results of this study provided important information on intrapulmonary PK of SPR206 in healthy subjects. Estimated intrapulmonary SPR206 penetration ratios determined from the ratio of AUC0-8 in ELF and AM to unbound plasma SPR206 concentrations were 0.264 and 0.328, respectively. Concentrations of SPR206 in ELF (range: 431.5 ng/mL to 735.5 ng/mL) and AM (range: 604.2 ng/mL to 860.6 ng/mL) were higher than the observed MIC90 of SPR206 against most contemporary Gram-negative organisms (≤ 500 ng/mL) based on data from in vitro and in vivo studies (26, 27, 30, 33, 39, 40). While ELF and AM concentrations were determined in healthy subjects in this study, intrapulmonary concentrations are expected to be higher in patients with respiratory infections, such as pneumonia (41). Administration of 3 doses of SPR206 100 mg IV q8h was well tolerated. Similar to the previous study, mostly mild paresthesias were the most common AE with no deaths or serious AEs and no clinically significant abnormalities observed for laboratory parameters, vital signs, ECG assessments, or physical examination.
Intrapulmonary concentrations of systemically administered antibiotics that exceed the MIC of target pathogens are important to achieve successful treatment outcomes from serious infections of the respiratory tract, especially those patients with pneumonia and in critical care units (37, 41–43). Beta-lactams, fluoroquinolones, macrolides, linezolid, and tigecycline demonstrate good penetration following oral or parenteral administration (41, 43, 44). In contrast, lung penetration with colistin and polymyxin B following IV administration is poor and highly variable. In animal models, lung concentrations of colistin were undetectable (45) or below the MIC50 for P. aeruginosa (46). Binding of colistin to mucin in airways results in subtherapeutic concentrations in the lung even with IV administration and could potentially promote development of resistance (47, 48). While data are limited for polymyxin B and colistin, IV administration of colistin in critically ill patients resulted in undetectable levels in BAL fluid (49), which may have been the result of extensive tissue binding (50). Additionally, achieving an adequate dose of polymyxin B or colistin to reach a bactericidal concentration in the target tissue can pose a challenge because of the potential for nephrotoxicity (46, 48, 51).
Serious infections due to MDR Gram-negative bacteria often are treated with aminoglycosides, carbapenems, and polymyxins. However, use of aminoglycosides and polymyxins is limited by serious side effects. Aminoglycosides and polymyxins are associated with an increased risk for nephrotoxicity (19, 52, 53). In addition, aminoglycosides increase the risk of ototoxicity, and polymyxins are associated with an increased risk for neurotoxicity (54). Both aminoglycosides and polymyxins require therapeutic drug monitoring to confirm that optimal drug concentrations are achieved and to avoid serious toxicity (55). Even at recommended dosages of polymyxins, acute kidney injury may occur in up to 60% of patients (56), and all-cause nephrotoxicity was reported in over 40% of patients and renal failure in 11.2% (18).
SPR206 is a novel polymyxin B analogue, which was developed with the goal of limiting the nephrotoxicity potential of polymyxins (34) while maintaining the in vitro and in vivo activity against MDR Gram-negative pathogens. Results from nonclinical studies showed that SPR206 reduced kidney cell cytotoxicity and had lower exposure in the kidney than polymyxin B (34, 35). These results together with results from Phase 1 studies (36, 57) suggest the potential of SPR206 to offer a broad spectrum of activity against MDR pathogens, but with an improvement in the safety profile compared with older polymyxins.
In summary, this study demonstrated that IV administration of SPR206 achieves plasma concentrations that were consistent with previous studies in healthy volunteers and exhibits lung penetration into ELF and AM. Concentrations of SPR206 in ELF and AM exceeded the MIC90 for most Gram-negative pathogens causing serious bacterial infections including A. baumannii, K. pneumoniae, and P. aeruginosa. These results support evaluation of SPR206 for treating serious respiratory infections caused by MDR Gram-negative pathogens.
MATERIALS AND METHODS
The study was conducted between May 2021 and September 2021 at the Medicines Evaluation Unit, Ltd., Manchester, United Kingdom in accordance with the U.S. Code of Federal Regulations and ethical principles of the Declaration of Helsinki, Good Clinical Practices, and the International Council for Harmonisation guidelines. The study protocol and all amendments were reviewed by the Institutional Review Board for the 2 study centers (NHS Health Research Authority, South Central - Berkshire Research Ethics Committee, Bristol, United Kingdom). Informed consent was obtained from each subject in writing before any study procedures were performed. This study was registered at clinicaltrials.gov: NCT04868292.
Study design.
This was a Phase 1, single-center, multiple-dose, open-label PK study in healthy adult male and female subjects. Subjects received three 100 mg doses of SPR206 administered q8h as a 1-h IV infusion. Three consecutive doses were sufficient to achieve steady-state based on a previous Phase 1 study in healthy subjects (36). A screening evaluation to determine eligibility for enrollment into the study was performed within 28 days of initial dosing. Subjects who met inclusion and exclusion criteria returned to the study unit 1 day before dosing (Day -1). Subjects who met inclusion and exclusion criteria were confined to the study site from Day -1 (1 day prior to start of dosing) and remained in the study site through completion of all scheduled procedures including collection of the last PK sample and bronchoscopy procedures along with safety evaluations (Day 2). Each subject underwent 1 bronchoscopy with BAL at 2, 3, 4, 6, or 8 h after the start of the third IV infusion. Subjects were required to return for a follow-up visit on Day 7 (+2 days) after discharge from the study site. The maximum duration of participation for each subject was up to 39 days (up to 28 days for Screening, 2 days of confinement, and up to 9 days for follow-up).
Study population.
Eligible subjects were adults aged 18 to 55 years with a body mass index (BMI) ≥18.5 and ≤32 kg/m2 and body weight between 55.0 and 100.0 kg who had been nonsmokers for at least 12 months prior to screening. Subjects were medically healthy without clinically significant abnormalities based on a screening medical history, physical examination, vital signs, 12-lead ECG, and clinical laboratory tests. Subjects had a forced expiratory volume in 1 s (FEV1) of at least 80% of predicted at screening; abstained from alcohol, caffeine, xanthine-containing beverages, or food for 48 h prior to and during the study; and agreed to use an effective method of contraception during the study.
At screening, subjects were excluded for a history of any significant medical condition; history (within 6 months) of known or suspected Clostridioides difficile infection; positive urine drug, alcohol, or cotinine testing; positive test for human immunodeficiency virus (HIV), hepatitis B surface antigen (HBsAg), or hepatitis C antibodies (HCV Ab); positive test for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); presence of fever, chills, or sweats; difficulty breathing; cough; sore throat; loss of taste or smell; or nausea, vomiting, or diarrhea or within 28 days prior to screening. Subjects with an ECG finding of QTcF interval duration ≥450 msec for males and 470 msec for females were excluded as were those with any clinical laboratory abnormalities.
Blood sample collection.
Blood samples for determining SPR206 plasma concentrations were collected within 60 min pre-dose (0 h) prior to second and third doses of SPR206 only and at 2, 3, 4, 6, and 8 h after the start of the third dose of SPR206.
Bronchoscopy and BAL.
Subjects were assigned to 1 of 5 bronchoscopy sampling times at 2, 3, 4, 6, or 8 (± 10 min) after the third IV infusion of SPR206. Each subject underwent 1 standardized bronchoscopy and BAL on Day 2 at the assigned time. A blood sample to determine plasma concentrations of SPR206 and urea was obtained during the bronchoscopy procedure at each BAL sampling time (approximating the time of collection of a second aspirate of BAL). Detail descriptions of the outpatient bronchoscopy, BAL procedures, and the handling, processing, and storage of samples have been previously described (58–60).
Determination of plasma concentrations of SPR206.
Concentrations of SPR206 in plasma were determined by a validated liquid chromatography with tandem mass spectrometry (LC-MS/MS) assay performed at QPS, LLC. For plasma, the assay range was 50 to 50,000 ng/mL. In plasma, intraday precision (% coefficient of variation) ranged from 1.3% to 7.7%, and accuracy (% relative error) ranged from -1.2% to 9.5%. Interday precision ranged from 2.9% to 5.3% and accuracy ranged from 1.6% to 6.0%.
Determination of BAL fluid and cell pellet concentrations of SPR206.
The concentrations of SPR206 in BAL and in cell pellets were determined by Keystone Bioanalytical, Inc. using a validated LC-MS/MS method. ELF concentrations were calculated by the urea dilution method (61, 62). AM concentrations were determined from cell pellet drug concentrations, cell count in BAL fluid, and macrophage cell volume (62). The calibration curves for SPR206 were linear over a range from 2 to 1,000 ng/mL. The inter-assay precision (%CV) and accuracy (%RE) for SPR206 were 2.81% to 15.84% and -4.02% to 1.51%, respectively. The intra-assay precision (%CV) and accuracy (%RE) for SPR206 were 1.43% to 18.59% and -17.44% to 6.28%, respectively.
Determination of urea concentration.
The concentration of urea in plasma and BAL fluid supernatants was determined using a validated LC/MS/MS method from Keystone Bioanalytical, Inc. Thirty human plasma and 30 BAL fluid samples were assayed for urea concentrations. A total of 10 samples for both plasma and BAL fluid (33.3%) were selected for testing, and the calculated assay variability was within 20%. The calibration curve for the urea assay in plasma was linear over the range from 100 to 3,000 μg/mL. The inter-assay precision (%CV) and accuracy (%RE) for SPR206 were 1.98% to 3.37% and -8.17% to 4.13%, respectively. The intra-assay precision (%CV) and accuracy (%RE) for SPR206 were 0.56% to 5.60% and -10.21% to 8.54%, respectively. The calibration range of the urea assay for BAL fluid was linear (r2 > 0.998) over the range of 0.2 to 10 μg/mL. The inter-assay precision (%CV) and accuracy (%RE) for SPR206 were 6.40% to 9.77% and -3.22% to 1.47%, respectively. The intra-assay precision (%CV) and accuracy (%RE) for SPR206 were 0.96% to 12.08% and -11.30% to 9.15%, respectively.
Pharmacokinetic analysis.
PK analysis of SPR206 plasma concentrations was determined with noncompartmental PK analysis using Phoenix WinNonlin software (version 8.3, Certara Inc.). PK parameters were Tmax, Cmax, Cmin, and AUC0-8.
ELF volume and antibiotic concentrations for ELF and AM.
Actual BAL samples were collected outside the protocol-specified sampling time windows. Thus, an updated method was utilized where the PK parameters for ELF or AM were calculated from composite concentration profiles of ELF or AM, respectively, based on the 27 subjects in the BAL PK population. Some blood samples were collected outside of the planned sampling windows, and therefore, individual plasma AUC0-8 was calculated first and the mean population plasma AUC0-8 was derived from all individual AUC0-8 values.
Concentrations of SPR206 were summarized separately for plasma, ELF, and AM by nominal time point using descriptive statistics. The mean concentrations of SPR206 in BAL fluid obtained at the different BAL fluid sampling times (e.g., 2, 3, 4, 6, and 8 h) were used to estimate the AUC0-8 for SPR206 in plasma, ELF, and AM. The concentration at the final sampling time (8 h) also served as the time zero value for determining the AUC0-8 value of each matrix using Phoenix WinNonlin software (version 8.3, Certara Inc.). The ratios of the AUC0-8 of ELF or AM to the AUC0-8 of plasma (total and unbound) were calculated. Plasma protein binding analyses were not conducted as part of this study. The value used for the unbound fraction of SPR206 in plasma was 0.914 (Spero Therapeutics, Inc., data on file). The measured concentrations in ELF and AM represented unbound concentrations, since only unbound plasma fractions are considered to penetrate the lung compartments. Individual plasma concentration-time profile versus time and arithmetic mean concentration (±SD) versus time plots were presented for the concentration-time data.
Calculations of the ELF volume and drug concentrations in ELF and AM were performed with BAL fluid supernatants and cell pellets from pooled aspirates (60). The concentration of SPR206 in ELF (CELF) was determined as follows: CELF = concentration in BAL × BAL volume/ELF volume. ELF volume was derived from ELF volume = BAL volume × ureaBAL/ureaplasma, where ureaBAL was the concentration of urea in BAL fluid and ureaplasma was the concentration of urea in plasma.
The concentration of SPR206 in AM (CAM) was determined using the equation CAM = Cpellet suspension/VAM, where Cpellet is the mass of SPR206 measured in the cell suspension and VAM is the volume of alveolar cells in the 1-mL cell suspension. A mean macrophage cell volume of 2.42 μL/106 cells was used for VAM (63, 64).
Statistical analysis.
The study was primarily a descriptive study comparing the steady-state concentrations of SPR206 in plasma, ELF, and AM at selected time intervals. Thus, no formal statistical hypothesis testing was performed. The sample size was based on the need to obtain adequate safety, tolerability, and PK data to achieve the objectives of the study while exposing as few subjects as possible to study medication and procedures. The sample size of 6 subjects per cohort was considered sufficient to provide adequate PK data at each BAL time point. SAS Version 9.4 was used to analyze the data, and create summary tables, subject data listings, and graphical representations of data.
Safety assessments.
Subjects were continuously monitored during the bronchoscopy. Vital signs (blood pressure, heart rate, and respiratory rate) were recorded prior to the scheduled bronchoscopy time and at 15 min and 1 h following the end of bronchoscopy. Subjects were kept under observation for 2 to 3 h post-bronchoscopy for safety evaluations. Continuous pulse oximetry was conducted from the beginning of preparation of the subject for bronchoscopy until the post-procedure oxygen saturation was at least 93% on room air. Pulse oximetry was extended if a clinically significant increase in heart rate or cardiac rhythm abnormalities were detected. Other safety assessments included physical examinations, clinical laboratory monitoring (hematology, blood chemistry, and urinalysis), 12-lead ECG, and adverse event recording.
ACKNOWLEDGMENTS
The authors acknowledge the editorial assistance of Richard S. Perry, PharmD in the preparation of this manuscript, which was supported by Spero Therapeutics, Inc., Cambridge, MA. This work was supported by the Office of the Assistant Secretary of Defense for Health Affairs, through the Joint Warfighter Medical Research Program under Award No. W81XWH 19 1 0295. Opinions, interpretations, conclusions, and recommendations are those of the author and are not necessarily endorsed by the Department of Defense.
All authors were involved in data analysis and interpretation, and all authors reviewed the manuscript for critical content and approved the final version for submission.
J.B. is a former employee of Spero Therapeutics, Inc. J.B.B. and K.H. are current employees of Spero Therapeutics, Inc. Keith A. Rodvold was a consultant to Spero Therapeutics, Inc.
REFERENCES
- 1.World Health Organization. 2017. WHO publishes list of bacteria for which new antibiotics are urgently needed. https://www.who.int/news/item/27-02-2017-who-publishes-list-of-bacteria-for-which-new-antibiotics-are-urgently-needed. Accessed 22 May 2023.
- 2.Centers for Disease Control. 2019. Antibiotic resistance threats in the United States, 2019. U.S. Department of Health and Human Services p. 4, CDC; Atlanta, GA. [Google Scholar]
- 3.Cai B, Echols R, Magee G, et al. 2017. Prevalence of carbapenem-resistant Gram-negative infections in the United States predominated by Acinetobacter baumannii and Pseudomonas aeruginosa. Open Forum Infect Dis 4:ofx176. doi: 10.1093/ofid/ofx176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Boral B, Unaldi Ö, Ergin A, Durmaz R, Eser ÖK, Acinetobacter Study Group . 2019. A prospective multicenter study on the evaluation of antimicrobial resistance and molecular epidemiology of multidrug-resistant Acinetobacter baumannii infections in intensive care units with clinical and environmental features. Ann Clin Microbiol Antimicrob 18:19. doi: 10.1186/s12941-019-0319-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bulens SN, Yi SH, Walters MS, Jacob JT, Bower C, Reno J, Wilson L, Vaeth E, Bamberg W, Janelle SJ, Lynfield R, Vagnone PS, Shaw K, Kainer M, Muleta D, Mounsey J, Dumyati G, Concannon C, Beldavs Z, Cassidy PM, Phipps EC, Kenslow N, Hancock EB, Kallen AJ. 2018. Carbapenem-nonsusceptible Acinetobacter baumannii, 8 US Metropolitan Areas, 2012–2015. Emerg Infect Dis 24:727–734. doi: 10.3201/eid2404.171461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gales AC, Seifert H, Gur D, Castanheira M, Jones RN, Sader HS. 2019. Antimicrobial susceptibility of Acinetobacter calcoaceticus-Acinetobacter baumannii complex and Stenotrophomonas maltophilia clinical isolates: results from the SENTRY Antimicrobial Surveillance Program (1997-2016). Open Forum Infect Dis 6:S34–S46. doi: 10.1093/ofid/ofy293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lynch JP, 3rd, Zhanel GG, Clark NM. 2017. Infections due to Acinetobacter baumannii in the ICU: treatment options. Semin Respir Crit Care Med 38:311–325. doi: 10.1055/s-0037-1599225. [DOI] [PubMed] [Google Scholar]
- 8.Cheng A, Chuang YC, Sun HY, Sheng WH, Yang CJ, Liao CH, Hsueh PR, Yang JL, Shen NJ, Wang JT, Hung CC, Chen YC, Chang SC. 2015. Excess mortality associated with colistin-tigecycline compared with colistin-carbapenem combination therapy for extensively drug-resistant Acinetobacter baumannii bacteremia: a multicenter prospective observational study. Crit Care Med 43:1194–1204. doi: 10.1097/CCM.0000000000000933. [DOI] [PubMed] [Google Scholar]
- 9.Clark NM, Zhanel GG, Lynch JP. 2016. Emergence of antimicrobial resistance among Acinetobacter species: a global threat. Curr Opin Crit Care 22:491–499. doi: 10.1097/MCC.0000000000000337. [DOI] [PubMed] [Google Scholar]
- 10.Cassini A, Högberg LD, Plachouras D, Quattrocchi A, Hoxha A, Simonsen GS, Colomb-Cotinat M, Kretzschmar ME, Devleesschauwer B, Cecchini M, Ouakrim DA, Oliveira TC, Struelens MJ, Suetens C, Monnet DL, Burden of AMR Collaborative Group . 2019. 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 19:56–66. doi: 10.1016/S1473-3099(18)30605-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Spellberg B, Bonomo RA. 2015. Combination therapy for extreme drug-resistant (XDR) Acinetobacter baumannii: ready for prime-time? Crit Care Med 43:1332–1334. doi: 10.1097/CCM.0000000000001029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wong D, Nielsen TB, Bonomo RA, Pantapalangkoor P, Luna B, Spellberg B. 2017. Clinical and pathophysiological overview of Acinetobacter infections: a century of challenges. Clin Microbiol Rev 30:409–447. doi: 10.1128/CMR.00058-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zilberberg MD, Nathanson BH, Sulham K, Fan W, Shorr AF. 2016. Multidrug resistance, inappropriate empiric therapy, and hospital mortality in Acinetobacter baumannii pneumonia and sepsis. Crit Care 20:221. doi: 10.1186/s13054-016-1392-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lemos EV, de la Hoz FP, Einarson TR, McGhan WF, Quevedo E, Castañeda C, Kawai K. 2014. Carbapenem resistance and mortality in patients with Acinetobacter baumannii infection: systematic review and meta-analysis. Clin Microbiol Infect 20:416–423. doi: 10.1111/1469-0691.12363. [DOI] [PubMed] [Google Scholar]
- 15.Mancuso G, Midiri A, Gerace E, Biondo C. 2021. Bacterial antibiotic resistance: the most critical pathogens. Pathogens 10:1310. doi: 10.3390/pathogens10101310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhang Y, Chen XL, Huang AW, Liu SL, Liu WJ, Zhang N, Lu XZ. 2016. Mortality attributable to carbapenem-resistant Pseudomonas aeruginosa bacteremia: a meta-analysis of cohort studies. Emerg Microbes Infect 5:e27. doi: 10.1038/emi.2016.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Thaden JT, Park LP, Maskarinec SA, Ruffin F, Fowler VG, Jr, van Duin D. 2017. Results from a 13-year prospective cohort study show increased mortality associated with bloodstream infections caused by Pseudomonas aeruginosa compared to other bacteria. Antimicrob Agents Chemother 61:e02671-16. doi: 10.1128/AAC.02671-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Falagas ME, Kyriakidou M, Voulgaris GL, Vokos F, Politi S, Kechagias KS. 2021. Clinical use of intravenous polymyxin B for the treatment of patients with multidrug-resistant Gram-negative bacterial infections: an evaluation of the current evidence. J Glob Antimicrob Resist 24:342–359. doi: 10.1016/j.jgar.2020.12.026. [DOI] [PubMed] [Google Scholar]
- 19.Nation RL, Rigatto MHP, Falci DR, Zavascki AP. 2019. Polymyxin acute kidney injury: dosing and other strategies to reduce toxicity. Antibiotics (Basel) 8:24. doi: 10.3390/antibiotics8010024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Amaya-Villar R, Garnacho-Montero J. 2019. How should we treat Acinetobacter pneumonia? Curr Opin Crit Care 25:465–472. doi: 10.1097/MCC.0000000000000649. [DOI] [PubMed] [Google Scholar]
- 21.De Oliveira DMP, Forde BM, Kidd TJ, Harris PNA, Schembri MA, Beatson SA, Paterson DL, Walker MJ. 2020. Antimicrobial resistance in ESKAPE pathogens. Clin Microbiol Rev 33:e00181-19. doi: 10.1128/CMR.00181-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sheu CC, Chang YT, Lin SY, Chen YH, Hsueh PR. 2019. Infections caused by carbapenem-resistant Enterobacteriaceae: an update on therapeutic options. Front Microbiol 10:80. doi: 10.3389/fmicb.2019.00080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.GBD 2019 Antimicrobial Resistance Collaborators. 2022. Global mortality associated with 33 bacterial pathogens in 2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 400:2221–2248. doi: 10.1016/S0140-6736(22)02185-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Piperaki ET, Tzouvelekis LS, Miriagou V, Daikos GL. 2019. Carbapenem-resistant Acinetobacter baumannii: in pursuit of an effective treatment. Clin Microbiol Infect 25:951–957. doi: 10.1016/j.cmi.2019.03.014. [DOI] [PubMed] [Google Scholar]
- 25.Abdul-Mutakabbir JC, Nguyen L, Stamper K, Maassen P, Lev K, Morrisette T, Kebriaei R, Rybak MJ. 2020. Activity of SPR206, a polymyxin derivative, compared to colistin alone and in combination against multidrug-resistant Pseudomonas aeruginosa strains. Open Forum Infect Dis 7:S662–S663. doi: 10.1093/ofid/ofaa439.1478. [DOI] [Google Scholar]
- 26.Arends SJR, Rhomberg PR, Lister T, Cotroneo N, Rubio A, Flamm RK, Mendes RE. 2018. Activity of an investigational polymyxin-b-like compound (SPR206) against a set of Enterobacteriaceae organisms responsible for human infections, poster #81. Abstr ESCMID-ASM.
- 27.Arends SJR, Rhomberg PR, Lister T, Cotroneo N, Rubio A, Flamm RK, Mendes RE. 2019. Activity of an investigational polymyxin-b-like compound (SPR206) against a set of gram-negative bacilli responsible for human infections, poster AAR-796. Abstr ASM Microbe.
- 28.Arends SJR, Rhomberg PR, Lister T, Cotroneo N, Rubio A, Flamm RK, Mendes RE. 2018. In vitro activity evaluation of a next-generation polymyxin, SPR206, against non-fermentative gram-negative bacilli responsible for human infections, poster #80. Abstr ESCMID-ASM.
- 29.Cotroneo N, Tomich A, Zou Y, Lister T, Brown P, Dawson M. 2019. In vitro bactericidal activity of next-generation polymyxin SPR206 against susceptible and multidrug-resistant (MDR) Acinetobacter baumannii, Pseudomonas aeruginosa, Escherchia coli, and Klebsiella pneumoniae as compared to levofloxacin and meropenem, poster AAR-797. Abstr ASM Microbe.
- 30.Zhang Y, Zhao C, Wang Q, Wang X, Chen H, Li H, Zhang F, Wang H. 2020. Evaluation of the in vitro activity of new polymyxin B analogue SPR206 against clinical MDR, colistin-resistant and tigecycline-resistant Gram-negative bacilli. J Antimicrob Chemother 75:2609–2615. doi: 10.1093/jac/dkaa217. [DOI] [PubMed] [Google Scholar]
- 31.Grosser L, Heang K, Teague J, Warn P, Corbett D, Dawson MJ, Rubio A. 2018. In vivo efficacy of SPR206 in murine lung and thigh infection models caused by multidrug resistant pathogens Pseudomonas aeruginosa and Acinetobacter baumanii, poster #139. Abstr ESCMID-ASM.
- 32.Grosser L, Lister T, Heang K, Brown P, Dawson M, Rubio A. 2019. In vivo efficacy of next-generation polymyxin SPR206 in an immunocompetent murine ascending UTI infection model caused by Escherichia coli, poster AAR-798. Abstr ASM Microbe.
- 33.Grosser L, Lister T, Heang K, Warn P, Corbett D, Dawson MJ, Rubio A. 2019. In vivo efficacy of SPR206 in murine lung and thigh infection models caused by multidrug resistant pathogens Pseudomonas aeruginosa and Acinetobacter baumannii, poster AAR-799. ASM Microbe.
- 34.Brown P, Abbott E, Abdulle O, Boakes S, Coleman S, Divall N, Duperchy E, Moss S, Rivers D, Simonovic M, Singh J, Stanway S, Wilson A, Dawson MJ. 2019. Design of next generation polymyxins with lower toxicity: the discovery of SPR206. ACS Infect Dis 5:1645–1656. doi: 10.1021/acsinfecdis.9b00217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Brown P, Boakes S, Duperchy E, Rivers D, Singh J, Dawson MJ. 2018. Understanding the SAR interplay for kidney exposure and cytotoxicity facilitates the design of improved polymyxin derivatives - Identification of SPR206 as a development candidate, poster #145. Abstr ESCMID-ASM.
- 36.Bruss J, Lister T, Gupta VK, Stone E, Morelli L, Lei Y, Melnick D. 2021. Single- and multiple-ascending-dose study of the safety, tolerability, and pharmacokinetics of the polymyxin derivative SPR206. Antimicrob Agents Chemother 65:e0073921. doi: 10.1128/AAC.00739-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Drwiega EN, Rodvold KA. 2022. Penetration of antibacterial agents into pulmonary epithelial lining fluid: an update. Clin Pharmacokinet 61:17–46. doi: 10.1007/s40262-021-01061-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rodvold KA, George JM, Yoo L. 2011. Penetration of ant-infection agents into pulmonary epithelial lining fluid: focus on antibacterial agents. Clin Pharmacokinet 50:637–664. doi: 10.2165/11594090-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 39.Mendes RE, Sader HS, Arends SJR, Cotroneo N, Critchley IA, Castanheira M. 2022. In vitro activity of SPR206 and comparator compounds against Enterobacterales isolates responsible for infections in United States hospitals. Open Forum Infect Dis 9(Suppl 2):1677. doi: 10.1093/ofid/ofac492.1307. [DOI] [Google Scholar]
- 40.Mendes RE, Safer HS, Arends SJR, Cotroneo N, Critchley IA, Castanheira M. 2022. Activity of SPR206 and comparator agents against Pseudomonas aeruginosa and Acinetobacter baumannii causing infections in United States hospitals. Open Forum Infect Dis 9(Suppl 2):1676. doi: 10.1093/ofid/ofac492.1306. [DOI] [Google Scholar]
- 41.Rodvold KA, Hope WW, Boyd SE. 2017. Considerations for effect site pharmacokinetics to estimate drug exposure: concentrations of antibiotics in the lung. Curr Opin Pharmacol 36:114–123. doi: 10.1016/j.coph.2017.09.019. [DOI] [PubMed] [Google Scholar]
- 42.Finazzi S, Luci G, Olivieri C, Langer M, Mandelli G, Corona A, Viaggi B, Di Paolo A. 2022. Tissue penetration of antimicrobials in intensive care unit patients: a systematic review-Part I. Antibiotics (Basel) 11:1164. doi: 10.3390/antibiotics11091164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Heffernan AJ, Sime FB, Lipman J, Dhanani J, Andrews K, Ellwood D, Grimwood K, Roberts JA. 2019. Intrapulmonary pharmacokinetics of antibiotics used to treat nosocomial pneumonia caused by Gram-negative bacilli: a systematic review. Int J Antimicrob Agents 53:234–245. doi: 10.1016/j.ijantimicag.2018.11.011. [DOI] [PubMed] [Google Scholar]
- 44.Lu Q, Girardi C, Zhang M, Bouhemad B, Louchahi K, Petitjean O, Wallet F, Becquemin M-H, La Naour G, Marquette C-H, Rouby J-J. 2010. Nebulized and intravenous colistin in experimental pneumonia caused by Pseudomonas aeruginosa. Intensive Care Med 36:1147–1155. doi: 10.1007/s00134-010-1879-4. [DOI] [PubMed] [Google Scholar]
- 45.Viaggi B, Cangialosi A, Langer M, Olivieri C, Gori A, Corona A, Finazzi S, Di Paolo A. 2022. Tissue penetration of antimicrobials in intensive care unit patients: a systematic review-Part II. Antibiotics (Basel) 11:1193. doi: 10.3390/antibiotics11091193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yapa SWS, Li J, Porter CJ, Nation RL, Patel K, McIntosh MP. 2013. Population pharmacokinetics of colistin methane sulfonate in rats: achieving sustained lung concentrations of colistin for targeting respiratory infections. Antimicrob Agents Chemother 57:5087–5095. doi: 10.1128/AAC.01127-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Huang JX, Blaskovich MA, Pelingon R, Ramu S, Kavanagh A, Elliott AG, Butler MS, Montgomery AB, Cooper MA. 2015. Mucin binding reduces colistin antimicrobial activity. Antimicrob Agents Chemother 59:5925–5931. doi: 10.1128/AAC.00808-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Michalopoulos AS, Falagas ME. 2011. Colistin: recent data on pharmacodynamics properties and clinical efficacy in critically ill patients. Ann Intensive Care 1:30. doi: 10.1186/2110-5820-1-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Imberti R, Cusato M, Villani P, Carnevale G, Iotti GA, Langer M, Regazzi M. 2010. Steady-state pharmacokinetics and bronchoalveolar lavage concentration of colistin in critically ill patients after intravenous colistin methanesulphonate administration. Chest 138:1333–1339. doi: 10.1378/chest.10-0463. [DOI] [PubMed] [Google Scholar]
- 50.Imberti R. 2010. Intravenous colistimethate administration and colistin lung tissue concentrations. Intensive Care Med 36:1795. doi: 10.1007/s00134-010-1960-z. [DOI] [PubMed] [Google Scholar]
- 51.Gurjar M. 2015. Colistin for lung infection: an update. J Intensive Care 3:3. doi: 10.1186/s40560-015-0072-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nang SC, Azad MAK, Velkov T, Zhou QT, Li J. 2021. Rescuing the last-line polymyxins: achievements and challenges. Pharmacol Rev 73:679–728. doi: 10.1124/pharmrev.120.000020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zavascki AP, Nation RL. 2017. Nephrotoxicity of polymyxins: Is there any difference between colistimethate and polymyxin B? Antimicrob Agents Chemother 61:e02319-16. doi: 10.1128/AAC.02319-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Aslan AT, Akova M, Paterson DL. 2022. Next-generation polymyxin class of antibiotics: a ray of hope illuminating a dark road. Antibiotics (Basel) 11:1711. doi: 10.3390/antibiotics11121711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jenkins A, Thomson AH, Brown NM, Semple Y, Sluman C, MacGowan A, Lovering AM, Wiffen PJ, (BSAC Working Party on Therapeutic Drug Monitoring) . 2016. Amikacin use and therapeutic drug monitoring in adults: do dose regimens and drug exposures affect either outcome or adverse events? A systematic review. J Antimicrob Chemother 71:2754–2759. doi: 10.1093/jac/dkw250. [DOI] [PubMed] [Google Scholar]
- 56.Tsuji BT, Pogue JM, Zavascki AP, Paul M, Daikos GL, Forrest A, Giacobbe DR, Viscoli C, Giamarellou H, Karaiskos I, Kaye D, Mouton JW, Tam VH, Thamlikitkul V, Wunderink RG, Li J, Nation RL, Kaye KS. 2019. International Consensus Guidelines for the Optimal Use of the Polymyxins: Endorsed by the American College of Clinical Pharmacy (ACCP), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), Infectious Diseases Society of America (IDSA), International Society for Anti-infective Pharmacology (ISAP), Society of Critical Care Medicine (SCCM), and Society of Infectious Diseases Pharmacists (SIDP). Pharmacotherapy 39:10–39. doi: 10.1002/phar.2209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bruss J, Gupta VK, Hamed K. 2023. Pharmacokinetics of SPR206, a semisynthetic polymyxin derivative, in subjects with varying degrees of renal impairment. Antimicrob Agents Chemother. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gotfried MH, Danziger LH, Rodvold KA. 2003. Steady-state plasma and bronchopulmonary characteristics of clarithromycin extended-release tablets in normal healthy adults. J Antimicrob Chemother 52:450–456. doi: 10.1093/jac/dkg355. [DOI] [PubMed] [Google Scholar]
- 59.Rodvold KA, Danziger LH, Gotfried MH. 2003. Steady-state plasma and bronchopulmonary concentrations of intravenous levofloxacin and azithromycin in healthy adults. Antimicrob Agents Chemother 47:2450–2457. doi: 10.1128/AAC.47.8.2450-2457.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rodvold KA, Gotfried MH, Danziger LH, Servi RJ. 1997. Intrapulmonary steady-state concentrations of clarithromycin and azithromycin in healthy adult volunteers. Antimicrob Agents Chemother 41:1399–1402. doi: 10.1128/AAC.41.6.1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Rennard SI, Basset G, Lecossier D, O'Donnell KM, Pinkston P, Martin PG, Crystal RG. 1986. Estimation of volume of epithelial lining fluid recovered by lavage using urea as marker for dilution. J Appl Physiol (1985) 60:532–538. doi: 10.1152/jappl.1986.60.2.532. [DOI] [PubMed] [Google Scholar]
- 62.Rodvold KA, Gotfried MH, Gupta V, Ek A, Srivastava P, Talley A, Bruss J. 2022. Plasma and intrapulmonary concentrations of tebipenem following oral administration of tebipenem pivoxil hydrobromide to healthy adult subjects. Antimicrob Agents Chemother 66:e0059022. doi: 10.1128/aac.00590-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Baldwin DR, Honeybourne D, Wise R. 1992. Pulmonary disposition of antimicrobial agents: methodological considerations. Antimicrob Agents Chemother 36:1171–1175. doi: 10.1128/AAC.36.6.1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Baldwin DR, Wise R, Andrews JM, Ashby JP, Honeybourne D. 1990. Azithromycin concentrations at the site of pulmonary infections. Eur Respir J 3:886–890. doi: 10.1183/09031936.93.03080886. [DOI] [PubMed] [Google Scholar]


