pulmonary pharmacokinetics of colistin following ... · pulmonary pharmacokinetics of colistin...

12
Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, a Qi Tony Zhou, b Yang Hu, c Nikolas J. Onufrak, d Siping Sun, a Jiping Wang, c Alan Forrest, d Hak-Kim Chan, a Jian Li e Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales, Australia a ; Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA b ; Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia c ; Department of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina, USA d ; Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia e ABSTRACT Colistin has been administered via nebulization for the treatment of re- spiratory tract infections. Recently, dry powder inhalation (DPI) has attracted increas- ing attention. The current study aimed to investigate the pharmacokinetics (PK) of colistin in epithelial lining fluid (ELF) and plasma following DPI and intravenous (i.v.) administration in healthy Sprague-Dawley rats. Rats were given colistin as DPI intra- tracheally (0.66 and 1.32 mg base/kg of body weight) or i.v. injection (0.66 mg base/ kg). Histopathological examination of lung tissue was performed at 24 h. Colistin concentrations in both ELF and plasma were quantified, and a population PK model was developed and compared to a previously published PK model of nebulized colistin in rats. A two-compartment structural model was developed to describe the PK of colistin in both ELF and plasma following pulmonary or i.v. administration. The model-estimated clearance from the central plasma compartment was 0.271 liter/h/kg (standard error [SE] 2.51%). The transfer of colistin from the ELF compartment to the plasma com- partment was best described by a first-order rate constant (clearance of colistin from the ELF compartment to the plasma compartment 4.03 10 4 liter/h/kg, SE 15%). DPI appeared to have a higher rate of absorption (time to the maximum concentra- tion in plasma after administration of colistin by DPI, 10 min) than nebulization (time to the maximum concentration in plasma after administration of colistin by nebulization, 20 to 30 min), but the systemic bioavailabilities by the two routes of administration were similar (46.5%, SE 8.43%). Histopathological examination re- vealed no significant differences in inflammation in lung tissues between the two treatments. Our findings suggest that colistin DPI is a promising alternative to nebulization considering the similar PK and safety profiles of the two forms of administration. The PK and histopathological information obtained is critical for the development of optimal aerosolized colistin regimens with activity against lung infections caused by Gram-negative bacteria. KEYWORDS polymyxin, colistin, dry powder, pulmonary delivery, disposition R espiratory tract infections caused by multidrug-resistant (MDR) Gram-negative bacteria are a major public health burden globally (1, 2). With their increasing prevalence and a scarcity of effective antibiotics, respiratory tract infections caused by these superbugs are alarmingly dangerous, often resulting in high rates of morbidity and mortality (3). Colistin (i.e., polymyxin E), a polypeptide antibiotic, has been increas- ingly used as a last resort for the treatment of respiratory tract infections caused by MDR Gram-negative pathogens (4, 5). Traditional parenteral administration of colistin Received 25 May 2017 Returned for modification 10 July 2017 Accepted 6 August 2017 Accepted manuscript posted online 14 August 2017 Citation Lin Y-W, Zhou QT, Hu Y, Onufrak NJ, Sun S, Wang J, Forrest A, Chan H-K, Li J. 2017. Pulmonary pharmacokinetics of colistin following administration of dry powder aerosols in rats. Antimicrob Agents Chemother 61:e00973-17. https://doi.org/10.1128/AAC .00973-17. Copyright © 2017 American Society for Microbiology. All Rights Reserved. Address correspondence to Hak-Kim Chan, [email protected], or Jian Li, [email protected]. Y.-W.L. and Q.T.Z. contributed equally to this article. PHARMACOLOGY crossm November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 1 Antimicrobial Agents and Chemotherapy on April 7, 2020 by guest http://aac.asm.org/ Downloaded from

Upload: others

Post on 21-Sep-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

Pulmonary Pharmacokinetics of Colistinfollowing Administration of Dry PowderAerosols in Rats

Yu-Wei Lin,a Qi Tony Zhou,b Yang Hu,c Nikolas J. Onufrak,d Siping Sun,a

Jiping Wang,c Alan Forrest,d Hak-Kim Chan,a Jian Lie

Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales,Australiaa; Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, WestLafayette, Indiana, USAb; Drug Delivery, Disposition, and Dynamics, Monash Institute of PharmaceuticalSciences, Monash University, Parkville, Victoria, Australiac; Department of Pharmacotherapy and ExperimentalTherapeutics, Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina,USAd; Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton,Victoria, Australiae

ABSTRACT Colistin has been administered via nebulization for the treatment of re-spiratory tract infections. Recently, dry powder inhalation (DPI) has attracted increas-ing attention. The current study aimed to investigate the pharmacokinetics (PK) ofcolistin in epithelial lining fluid (ELF) and plasma following DPI and intravenous (i.v.)administration in healthy Sprague-Dawley rats. Rats were given colistin as DPI intra-tracheally (0.66 and 1.32 mg base/kg of body weight) or i.v. injection (0.66 mg base/kg). Histopathological examination of lung tissue was performed at 24 h. Colistinconcentrations in both ELF and plasma were quantified, and a population PK model wasdeveloped and compared to a previously published PK model of nebulized colistin inrats. A two-compartment structural model was developed to describe the PK of colistinin both ELF and plasma following pulmonary or i.v. administration. The model-estimatedclearance from the central plasma compartment was 0.271 liter/h/kg (standard error[SE] � 2.51%). The transfer of colistin from the ELF compartment to the plasma com-partment was best described by a first-order rate constant (clearance of colistin from theELF compartment to the plasma compartment � 4.03 � 10�4 liter/h/kg, SE � 15%).DPI appeared to have a higher rate of absorption (time to the maximum concentra-tion in plasma after administration of colistin by DPI, �10 min) than nebulization(time to the maximum concentration in plasma after administration of colistin bynebulization, 20 to 30 min), but the systemic bioavailabilities by the two routes ofadministration were similar (�46.5%, SE � 8.43%). Histopathological examination re-vealed no significant differences in inflammation in lung tissues between the twotreatments. Our findings suggest that colistin DPI is a promising alternative tonebulization considering the similar PK and safety profiles of the two forms ofadministration. The PK and histopathological information obtained is critical forthe development of optimal aerosolized colistin regimens with activity againstlung infections caused by Gram-negative bacteria.

KEYWORDS polymyxin, colistin, dry powder, pulmonary delivery, disposition

Respiratory tract infections caused by multidrug-resistant (MDR) Gram-negativebacteria are a major public health burden globally (1, 2). With their increasing

prevalence and a scarcity of effective antibiotics, respiratory tract infections caused bythese superbugs are alarmingly dangerous, often resulting in high rates of morbidityand mortality (3). Colistin (i.e., polymyxin E), a polypeptide antibiotic, has been increas-ingly used as a last resort for the treatment of respiratory tract infections caused byMDR Gram-negative pathogens (4, 5). Traditional parenteral administration of colistin

Received 25 May 2017 Returned formodification 10 July 2017 Accepted 6August 2017

Accepted manuscript posted online 14August 2017

Citation Lin Y-W, Zhou QT, Hu Y, Onufrak NJ,Sun S, Wang J, Forrest A, Chan H-K, Li J. 2017.Pulmonary pharmacokinetics of colistinfollowing administration of dry powderaerosols in rats. Antimicrob Agents Chemother61:e00973-17. https://doi.org/10.1128/AAC.00973-17.

Copyright © 2017 American Society forMicrobiology. All Rights Reserved.

Address correspondence to Hak-Kim Chan,[email protected], or Jian Li,[email protected].

Y.-W.L. and Q.T.Z. contributed equally to thisarticle.

PHARMACOLOGY

crossm

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 1Antimicrobial Agents and Chemotherapy

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 2: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

may not provide optimal efficacy for the treatment of respiratory tract infections, asrecent animal studies demonstrated the very limited exposure of colistin in theepithelial lining fluid (ELF) (6–10). A recent pharmacokinetic/pharmacodynamic (PK/PD)study of parenteral colistin against Pseudomonas aeruginosa in mouse thigh and lunginfection models was conducted; the ratio of the area under the unbound (free) plasmaconcentration-time curve over the MIC (fAUCplasma/MIC) required to achieve bacterios-tasis in the lungs was approximately 2.5- to 5-fold higher than that required to achievebacteriostasis in the thighs (11). Increasing the dose of parenteral colistin is not feasibledue to the dose-limiting nephrotoxicity of colistin (12, 13), which highlights the needfor alternative routes of colistin administration to treat respiratory tract infections.

Pulmonary administration of colistin can achieve a high drug exposure in the lungswhile minimizing systemic exposure and nephrotoxicity. The advantage of the pulmo-nary delivery of colistin has been illustrated in several preclinical (6–10, 14) and clinical(15, 16) PK studies. Yapa et al. reported in a clinical study that pulmonary nebulizationof colistin methanesulfonate (CMS) (2 million and 4 million international units oncedaily, equivalent to 60 and 120 mg colistin base activity [CBA], respectively) resulted inhigher exposure of CMS and formed colistin in sputum (maximum sputum concentra-tion [Cmax, sputum] � 2 to 21 mg/liter) than that achieved after intravenous (i.v.)administration (Cmax, sputum � 0.12 to 0.72 mg/liter) (15). In a PK/PD study of colistinagainst P. aeruginosa ATCC 27853 in the mouse lungs, the required plasma fAUCplasma/MIC following pulmonary administration (2.99) was 11-fold lower than that followingsubcutaneous administration (34.1) (11, 17). Nebulized antimicrobials have improvedthe life expectancy of patients with cystic fibrosis (CF) (18–20); however, they requireexpensive and complicated delivery devices and prolonged administration times (21–27), and the delivery efficiency is low (�15% of the administered dose reaches thelungs following jet nebulization) (28). Consequently, patient compliance with nebulizedantibiotics is low (29, 30). Dry powder inhalation (DPI) provides a suitable alternative, aspowder inhaler devices are portable, easy to use, and, most importantly, able to deliverdrug efficiently (31–33) and improve efficacy and patient compliance (34, 35).

To date, the majority of in vivo studies of aerosolized CMS and colistin have focusedon nebulization (6–10, 14, 15, 36–39), whereas only a few investigated DPI (23–26). Thesafety and efficacy of jet-milled CMS/colistin powder have been evaluated in healthyvolunteers and patients with CF (23–26). However, these DPI formulations have arelatively low aerosolization efficiency (40% in the fine-particle fraction [FPF]) (23). Inour earlier study, inhalable dry powder formulations of colistin with a high aerosoliza-tion efficiency were developed by spray-drying (total FPF, �83% via an Aerolizerdevice) (22). These highly dispersible colistin DPI formulations provide higher levels ofdrug deposition in the lungs and thus have a greater potential for the effectivetreatment of respiratory infections caused by MDR Gram-negative bacteria (40, 41).Although a CMS DPI product (Colobreathe) has been approved in Europe, the dosageregimens of DPI have not been appropriately optimized on the basis of PK/PD studies(32). The present study aimed to investigate the PK of our highly efficient colistin DPIformulation in healthy rats after pulmonary administration.

RESULTS

Following DPI, colistin was rapidly absorbed into the systemic circulation, with themaximum plasma concentration (Cmax, plasma) being detected within 10 min afteradministration, followed by a biexponential decline over the 12-h sampling period (Fig.1 and Table 1). High levels of colistin exposure in the ELF were achieved following DPI(maximum ELF concentration [Cmax, ELF] � 400.1 � 44.9 and 607.0 � 58.9 mg/liter fordoses of 0.66 and 1.32 mg colistin base/kg of body weight, respectively; Table 1) andwas maintained for at least 12 h (Fig. 1). The concentrations of urea measured in thebronchoalveolar lavage fluid (BALF) and plasma were 1.01 � 0.43 mg/dl and 48 � 10mg/dl, respectively.

The disposition of colistin in the ELF and plasma following DPI and i.v. administra-tion was best described by two compartments (Fig. 2 to 4 ; see also Fig. S1 in the

Lin et al. Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 2

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 3: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

supplemental material). The PK parameters generated from the population PK modelare listed in Table 2. The estimated apparent volumes of distribution in the ELF (VELF)in ELF compartments 1 and 2 (VE1 and VE2, respectively; manifested as ELF1 and ELF2in Fig. 4, respectively) were 2.90 � 10�5 liter/kg (standard error [SE] � 35.7%) and9.67 � 10�4 liter/kg (SE � 18.8%), respectively. The estimated total systemic bodyclearance (CLtotal) from the central plasma compartment for colistin was 0.271 liter/h/kg(SE � 2.51%). A first-order process was sufficient to describe the transfer of colistin fromELF to the central plasma compartment. The estimated systemic bioavailability of DPI(FDPI) was 46.5% (SE � 8.43%) (Table 2).

Histopathological examination of the lung tissue at 24 h following DPI and wet

FIG 1 (A) Colistin ELF concentration-time profiles following DPI of 0.66 mg base/kg and 1.32 mg base/kgcolistin. The colistin ELF concentration following i.v. bolus administration was below the LOQ. (B) Totalplasma colistin concentration-time profiles following DPI and i.v. administration of colistin in healthySprague-Dawley rats. Each symbol represents the mean � standard deviation (SD; n � 3 or more). *, theconcentration is below the LOQ (0.10 mg/liter).

TABLE 1 PK parameters for colistin in healthy rats following DPI administrationa

DoseCmax, ELF

(mg/liter)Cmax, plasma

(mg/liter) Tmax (min)

Single pulmonary dose of:0.66 mg base/kg 400.1 � 44.9 0.65 � 0.32 101.32 mg base/kg 607.0 � 58.9 1.66 � 0.69 10

Single i.v. dose of 0.66 mg base/kg �LOQ NA NAaData are presented as means � standard deviations. NA, not applicable.

PK of Aerosolized Colistin Dry Powder Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 3

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 4: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

FIG 2 Visual predictive checks for colistin in plasma following DPI of 0.66 mg base/kg (A) and 1.32 mgbase/kg (B) and i.v. administration of 0.66 mg base/kg (C). Solid line, the median model-predictedconcentrations (P50); broken lines, model-predicted 10th percentile (P10) and 90th percentile (P90)concentrations; solid dots, observed concentrations. After 4 h, the 10th percentile concentration ap-proaches zero. *, the concentration is below the LOQ (0.10 mg/liter).

Lin et al. Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 4

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 5: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

nebulization of colistin showed mean pathological scores of about 1.0 to �2.0 (indi-cating mild damage), whereas the saline-treated group had a mean pathological scoreof 0.3 (indicating no damage; Table 3). Furthermore, no significant difference in thepathological scores was observed among the lung tissue samples obtained at 24 h fromthe nebulization (1.32 mg base/kg) and DPI (0.66 mg base/kg) groups (Table 3). Evenwith the highest dose (1.32 mg base/kg), DPI and wet nebulization resulted in milddamage in the lung tissue with similar pathological scores (1.9 and 1.5, respectively)(Table 3).

DISCUSSION

Aerosolized polymyxin therapy administered via nebulization has become a com-mon complementary practice for the treatment of respiratory tract infections, such asthose in CF patients (42, 43) and in critically ill patients with ventilator-associatedpneumonia (44, 45). With recent advancements in particle engineering technology, wehave successfully formulated a colistin dry powder with a markedly higher aerosoliza-tion efficiency (total FPF, �83% via an Aerolizer device) than those colistin dry powderformulations in the previous studies (22, 25, 26). DPI offers a potentially superioralternative inhalational delivery method that provides a better drug deposition in thelungs than nebulization and that results in a higher rate of patient compliance than

FIG 3 Visual predictive checks for colistin in ELF following DPI of 0.66 mg base/kg (A) and 1.32 mgbase/kg colistin (B). Solid line, the median model-predicted concentrations (P50); broken lines, themodel-predicted 10th percentile (P10), and 90th percentile (P90) concentrations; solid dots, observedconcentrations.

PK of Aerosolized Colistin Dry Powder Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 5

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 6: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

nebulization (23–26, 33). This is the first preclinical study to investigate the PK of thespray-dried colistin powder aerosol in healthy rats.

In the present study, a population PK model was constructed to better understandthe disposition of colistin in ELF and plasma following pulmonary and i.v. administra-tion. Data were distributed around the line of identity (see Fig. S1 in the supplementalmaterial), and the majority of the observed concentrations were scattered within the10th and 90th percentiles (Fig. 2 and 3). Therefore, our population PK model welldescribed colistin exposure in ELF (overall R2 � 0.87) and plasma (overall R2 � 0.91).The model revealed that the plasma concentrations of colistin rapidly decreasedfollowing i.v. and pulmonary administration (Fig. 1). Following administration of colis-tin, the model-predicted total systemic body clearance (CLtotal � 0.271 liter/h/kg, SE �

2.51%; Table 2) and volume of distribution of the central plasma compartment (Vc �

TABLE 2 Estimated values of population PK parameters for colistin in healthy rats following DPI or i.v. administration

Parameter Description Unit Estimated value SE (%)

VE1 Vol of distribution in ELF1 (ELF compartment) liter/kg 2.90 � 10�5 35.7VE2 Vol of distribution in ELF2 (peripheral lung compartment) liter/kg 9.67 � 10�4 18.8Vc Volume of distribution in central plasma compartment liter/kg 0.572 17.9Vp Volume of distribution in peripheral plasma compartment liter/kg 1.47 35.3CLD, ELF ELF intercompartmental clearance liter/h/kg 4.26 � 10�4 37.8CLD, plasma Plasma intercompartmental clearance liter/h/kg 0.172 24.3CLtotal Total systemic body clearance liter/h/kg 0.271 2.51CLELF, plasma Intercompartmental clearance from ELF to plasma liter/h/kg 4.03 � 10�4 15.0CLplasma, ELF Intercompartmental clearance from plasma to ELF liter/h/kg 9.93 � 10�7 46.0fu Unbound fraction 0.44 (fixed) NAa

FDPI Bioavailability % 46.5 8.43aNA, not applicable.

FIG 4 A population PK structural model describing the disposition of colistin in ELF and plasma followingDPI or i.v. administration of colistin. The parameters are presented in Table 2.

Lin et al. Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 6

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 7: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

0.572 liter/kg, SE � 17.9%; Table 2) were comparable to those reported in the literature(Vc � 0.22 to 0.43 liter/kg and CLtotal � 0.22 to 0.43 liter/h/kg) (7, 10, 36, 46).

In the current study, the PK linearity observed with colistin following DPI adminis-tration in healthy Sprague-Dawley rats is consistent with the findings of Yapa et al.following wet nebulization in rats (7). In contrast, PK nonlinearity was reported byGontijo et al. following wet nebulization of colistin in rats (10). A more complex PKmodel with Michaelis-Menten (nonlinear) kinetics was proposed by Gontijo et al. todescribe the transfer of colistin across the lung alveolar epithelium after nebulization(10). However, we did not find a significant improvement in model fit upon incorpo-ration of such a process, with a first-order rate constant being sufficient to describe ourdata. This is in agreement with the PK models developed for rats (6), sheep (14), baboonmonkeys (9), and critically ill patients (16) after the nebulization of colistin. Themechanism of colistin transport across the lung epithelium remains unclear and isbeing investigated in our laboratory.

The systemic absorption of colistin was faster in rats when it was administered asDPI, with the time to reach the maximum plasma concentration (Tmax) being 20 to 30min for the nebulization and �10 min for DPI (7). The slightly faster absorption ratefollowing DPI is very likely due to the higher aerosolization efficiency of spray-driedcolistin powder and the differential distribution of PEPT2 in different regions of therespiratory tract (47, 48). As opposed to wet nebulization using a MicroSprayer device,the large fine-particle fraction of the colistin dry powder upon pulmonary administra-tion is able to reach deeper airways, where PEPT2 is abundantly expressed (47–49).Such a faster absorption rate with DPI than with nebulization was also observed in ourearlier study, where the Kv1.3-blocking peptide HsTX1[R14A] was administered tohealthy rats using the same Penn-Century device used in the present study (50).Despite the more rapid absorption of the powder formulation, the systemic bioavail-ability of colistin solution (Fsolution � 31 to 69%) (7, 10) and the spray-dried powderformulation (FDPI � 46.5%) (Table 2) was similar in healthy Sprague-Dawley rats. Thetranslation of PK data from rats to humans after pulmonary administration has re-mained challenging, as the airway anatomy of rats differs from that of humans (51) andthe Penn-Century device dispersed powder differently from the clinical dry powderinhaler (52). With the Penn-Century device, the colistin dry powder is dispersedpassively into the lungs, which may not be comparable to the dispersal achieved withclinically used dry powder inhalers (i.e., Twincer), with which patients are required toactively inhale CMS/colistin dry powder (25–27). Importantly, the human PK of CMSadministered via clinical dry powder inhalers are likely influenced by interindividual andinterdevice variabilities (27). Clinical studies are thus needed to characterize the extentof potential patient- and device-specific differences in the PK of DPI of polymyxins.

Consistent with the observation following the nebulization of colistin, DPI treatmentresulted in the prolonged and extensive exposure of colistin in the ELF (Fig. 1) (6, 7, 10,36). The colistin exposure in the ELF achieved after DPI (0.35 mg base/kg colistin),reflected by an area under the concentration-time curve (AUC) from 0 to 4 postdosingfor ELF (AUC0 – 4 h,ELF; �328 mg · h/liter, calculated using PK simulation), was compa-

TABLE 3 Histopathological examination of the lungs of healthy rats following DPI ofcolistin

Dose(mg base/kg) Formulation

Time(h)

No. ofrats

Mean pathologicalscore

Saline Solution 0.5 1.0Saline Solution 24 0.30.66 Dry powder 0.5 4 1.50.66 Dry powder 24 4 1.01.32 Dry powder 0.5 4 1.81.32 Dry powder 2 4 1.21.32 Dry powder 6 4 0.81.32 Dry powder 24 4 1.91.32 Solution 24 4 1.5

PK of Aerosolized Colistin Dry Powder Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 7

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 8: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

rable to that following wet nebulization of 0.35 mg base/kg colistin in healthy rats(AUC0 – 4 h,ELF � �369 mg · h/liter) (10). It is important to appreciate that ELF exposurecan be influenced by VELF, which was variable in rat studies (7.6 � 5.8 to 110 � 23 �l)due to the different techniques used for the sampling of BALF and the urea quantifi-cation method utilized (6, 7, 10, 36).

A key finding of our current study is that colistin concentrations in ELF were muchhigher following DPI treatment (Cmax, ELF � 400.1 � 44.9 and 607.0 � 58.9 mg/liter forcolistin at 0.66 and 1.32 mg base/kg, respectively) than following i.v. administration,with the concentrations dramatically exceeding the MIC90 (�1 mg/liter) against P.aeruginosa for at least 12 h (Fig. 2) (53). Colistin ELF concentrations following i.v.administration were below the limit of quantitation (LOQ) of ELF (4.84 mg/liter, afterconsidering the dilution factor for the interconversion of BALF and ELF concentrations).The population PK model developed in our study facilitated the prediction of colistinconcentrations in ELF following i.v. administration. Our results show that the simulatedcolistin concentration in ELF after i.v. administration was well below the ELF LOQ(�4.84 mg/liter) at any time point. This was consistent with the observation in patientswith CF, in which a low concentration of formed colistin (�1 mg/liter) was observedfollowing i.v. administration of CMS (150 mg CBA) (15). Another important finding inour population PK model is that the model-estimated VE2 was larger than the model-estimated VE1 (Table 2). This potentially infers the binding of colistin to pulmonarysubstances, such as mucin (54) and pulmonary surfactant (55), or a significant distri-bution of colistin into lung epithelial cells in rats. This finding is consistent with theliterature that colistin accumulates in the lung tissue (56) and binds to lung epitheliumvia an electrostatic interaction (57).

Collectively, the population PK model developed in the current study suggests thatthe spray-dried colistin DPI formulation has PK properties (e.g., ELF exposure andsystemic bioavailability) comparable to those of the wet nebulized formulation. Themajority of CF patients usually require long-term nebulized colistin therapy (as CMS)(58). Although nebulized colistin therapy has significantly improved patients’ lifeexpectancy, the complex nebulization system may have a negative impact on thequality of life and compliance in patients (27). On the basis of the findings of previousclinical studies (59, 60), the inhalation of colistin via dry powder inhalers is an attractiveand effective alternative for the treatment of respiratory infections owing to theimproved convenience, reduced duration of administration, and positive effects onquality of life, compliance, and, possibly, therapeutic outcomes in patients (61).

Aerosolized colistin in both solution and dry powder forms may cause pulmonaryadverse effects, such as a cough or throat irritation (32). The mechanisms underlyingsuch effects are unclear but may be related to mast cell degranulation (62, 63). Thesafety of pulmonary administration of colistin solution was previously examined, andno significant inflammation or damage to the lungs was observed after a single doseof CMS or colistin in rats (the doses were not provided) (6, 7). In the present study, thesafety of colistin DPI at 0.66 and 1.32 mg base/kg was evaluated by histopathologicalexamination and compared to the findings obtained following a similar dose of colistinsolution (Table 3). The pulmonary administration of neither colistin solution nor DPIresulted in severe inflammation or damage to the lung epithelium (scores, 1.0 to �2.0,showing mild to moderate lesions). Our results indicate that DPI of colistin is as safe asnebulization in healthy rats. PK/PD/toxicodynamic optimization of aerosolized poly-myxins is needed in patients to minimize any potential side effects (i.e., bronchospasmand cough).

To the best of our knowledge, the PK model described here is the first detailedpreclinical PK model describing the disposition of colistin in ELF and plasma followingDPI in rats. Our study has shown that spray-dried colistin DPI achieves drug exposurein ELF dramatically higher than that achieved by i.v. administration. Furthermore,histopathological examination of lung tissue demonstrates that DPI has safety compa-rable to that of wet nebulization. DPI is a promising alternative to nebulization,especially considering the convenience of the dry powder inhaler and its positive

Lin et al. Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 8

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 9: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

impacts on patient compliance. With the increasing incidence of life-threatening lunginfections caused by MDR Gram-negative bacteria, the PK and safety informationobtained here is critical to the optimization of DPI of polymyxin therapy in patients.

MATERIALS AND METHODSChemicals. Colistin sulfate (lot number 08M1526V; �15,000 U/mg) was purchased from Sigma-

Aldrich (St. Louis, MO, USA). All organic solvents were of analytical grade unless stated otherwise.Colistin dry powder. Colistin dry powder was prepared by spray-drying as previously described (22).

Briefly, colistin sulfate was dissolved in water and spray-drying was performed using a B-290 mini-spraydryer (Buchi Laboratories, Flawil, Switzerland). The spray-drying conditions were as follows: inlet tem-perature, 80°C; atomizer setting, 700 liter/h; aspirator, 40 m3/h; and feed rate, 2 ml/min. The spray-driedsamples were stored in a desiccator at room temperature.

Animals. All rat experiments (see Table S1 in the supplemental material) were approved by theAnimal Ethics Committee of the Monash Institute of Pharmaceutical Sciences, Monash University, andconducted in accordance with the Australian Code of Practice for the Care and Use of Animals forScientific Purposes. Male Sprague-Dawley rats (weight, 300 to 350 g; Monash Animal Research Platform,Victoria, Australia) were housed with a 12-h light/12-h dark cycle at ambient temperature (21 � 3°C) anda relative humidity of 40 to 70%. Food and water were available ad libitum.

Pharmacokinetic studies. Prior to the PK studies, the right carotid artery and right jugular vein ofrats were cannulated for blood collection and i.v. drug administration, respectively (6, 7). Followingsurgery, the rats were individually housed in metabolic cages and allowed to recover overnight.

Dosing of colistin. A single-dose PK study was performed in healthy rats following administration ofan i.v. bolus of colistin at 0.66 mg base/kg via the right jugular vein (n � 3). Blood samples were collectedat 10, 20, and 30 min and 1, 1.5, 2, 3, 4, 6, 12, and 24 h postadministration. Extra rats (n � 3 per time point)received an i.v. bolus of colistin at 0.66 mg base/kg for BALF collection (6, 7) at 10 min and 2, 6, and 12h postadministration.

DPI of colistin. Two groups of rats were utilized to characterize the pulmonary and systemic PK ofcolistin following DPI. In group 1, rats were given a colistin powder formulation (0.66 and 1.32 mgbase/kg) by pulmonary administration. Briefly, rats were anesthetized using gaseous isoflurane (AbbottAnimal Health, Abbott Park, IL). The anesthetized rats were placed on a Perspex support in a verticalupright position. The pulmonary administration was performed using a dry powder insufflator (DP-4M;Penn-Century, Glenside, PA) (52). The chamber of the dry powder insufflator was loaded with colistin drypowder, and 6 puffs of 2 ml air were applied via an air pump (AP-1; Penn-Century, Glenside, PA) todisperse the powder. The total drug administration time was approximately 20 to 30 s per animal. Thedry powder insufflator was weighed before and after administration to determine the amount of colistindelivered. On average, more than 90% of the colistin powder was dispersed and emitted from the device.The insufflator was cleaned with 10 puffs of 3 ml air before and after each experiment to remove excesspowder in the chamber and cannula. Blood samples were collected via the right carotid artery at 10 and30 min and 1, 2, and 4 h after pulmonary administration. In group 2, rats were given colistin dry powder(0.66 and 1.32 mg base/kg) by pulmonary administration, and BALF was collected at 10 and 30 min and1, 2, 4, 6, and 12 h (6, 7). In group 1, at least 3 animals were included per dosage regimen, while in group2, at least 3 animals were employed per time point.

The maximum dosage regimens for i.v. and pulmonary administration of the colistin dry powderaerosol were chosen on the basis of its tolerability in rats (6). The lowest dose selected was based uponthe LOQ of the method used for the analysis of colistin in plasma.

Measurement of colistin concentrations in plasma, BALF, and ELF. Colistin concentrations inplasma and BALF samples were determined by a validated liquid chromatography-mass spectrometry(LC-MS) method (64), with minor modifications. Briefly, BALF and plasma samples were deproteinizedwith 0.1% formic acid in acetonitrile (1:2 dilution) and centrifuged at 18,210 � g. For each analytical run,peak area ratios of the analyte (the sum of the peak areas for colistin A and colistin B) and the internalstandard (the sum of the peak areas for polymyxin B1 and polymyxin B2) were plotted against thenominal concentrations of the calibration standards. Linear least-squares regression analysis withoutweighting was performed to determine the linearity, intercept, and slope. Calibration curves wereprepared with blank BALF or plasma, and the concentrations ranged from 0.10 to 10.0 mg/liter. The LOQwas 0.10 mg/liter for both plasma and BALF samples. The inter- and intraday accuracy and precision ofthe assay for plasma and BALF samples are provided in Table S2. The VELF was determined using urea asan endogenous dilution marker (65). The urea concentrations in BALF and plasma were determined usinga QuantiChrom urea assay kit (Bioassay Systems, CA, USA). The standard curve was prepared with ureain Milli-Q water, and the linear detection range was from 0.08 mg/dl (13 �M) to 100 mg/dl (17 mM) urea.The accuracy and precision of the QuantiChrom urea assay kit for the measurement of the concentrationof urea in biological samples were examined previously (7). Quality control samples were prepared at 4levels, and the accuracy and precision of the kit at the 4 levels (1, 10, 25, and 50 mg/dl) were within 10%.

VELF was calculated as follows: VELF � ([urea]BALF/[urea]plasma) · VBALF, where [urea]BALF and [urea]plasma

are the urea concentrations (in milligrams per deciliter) in BALF and plasma, respectively, and VBALF is thevolume of BALF recovered.

The colistin concentration in ELF was calculated as follows: [colistin]ELF � [colistin]BALF · ([urea]plasma/[urea]BALF), where [colistin]ELF and [colistin]BALF are the colistin concentrations (in milligrams per liter) inELF and BALF, respectively.

PK of Aerosolized Colistin Dry Powder Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 9

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 10: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

Population pharmacokinetic model. A nonlinear mixed-effects model was employed to character-ize the disposition of colistin in ELF and plasma following DPI in health rats. During PK analysis, severalstructural models were tested and evaluated using the Monte Carlo parametric expectation maximizationalgorithm (importance sampling, pmethod � 4) in the S-ADAPT program (version 1.57), facilitated by theS-ADAPT TRAN set of programs (66). In the final composite model, two compartments were required todescribe the kinetics of colistin in both ELF and plasma following pulmonary and i.v. administration. Theresidual error model was estimated using heteroscedastic error (additive and proportional) structures.The Beal M3 method was employed to handle concentrations below the LOQ (67). Graphical analysis ofgoodness-of-fit plots (e.g., observed concentration versus population predicted concentration) andobjective function values (reported as �1 � log likelihood in S-ADAPT) was employed to evaluate thesuitability of the model (68–70). A decrease in the objective function value of 1.92 units (chi-square testwith 1 degree of freedom) was considered significant. The final model was evaluated using the visualpredictive checks (71).

Histopathological examination of the lungs following treatment with aerosolized colistin.Histopathological examination of rat lung tissue was performed to compare the effect of colistin onpulmonary epithelial cells after the pulmonary administration of colistin by DPI and by nebulization.Treatment groups received either 0.66 or 1.32 mg base/kg colistin dry powder or 1.32 mg base/kg colistinsolution, and the control group received 50 �l saline. Rats were humanely killed at 0.5 and 24 h (0.66 mgbase/kg) or at 0.5, 2, 6, and 24 h (1.32 mg base/kg) following dosing. To avoid any artificial damage,bronchoalveolar lavage and cardiac puncture were not performed prior to tissue harvesting. To quantifythe extent of lung damage, a pathological grading system developed by the Australian PhenomicsNetwork was adapted (Australian Phenomics Network Histopathology and Organ Pathology Service,Victoria, Australia). Briefly, the severity and the nature of the histopathological changes in lung epithelialcells were graded as follows: score of 0, no changes or mild changes considered insignificant; score of1, mild damage; score of �2, mild to moderate damage; and score of �3, moderate to severe damage.Scoring and grading of the lungs were performed blind by histopathologists at the Australian PhenomicsNetwork Histopathology and Organ Pathology Service.

SUPPLEMENTAL MATERIAL

Supplemental material for this article may be found at https://doi.org/10.1128/AAC.00973-17.

SUPPLEMENTAL FILE 1, PDF file, 0.3 MB.

ACKNOWLEDGMENTSWe acknowledge the financial support from a National Health and Medical Research

Council (NHMRC) project grant (APP1065046). J.L. and A.F. are supported by a researchgrant from the National Institute of Allergy and Infectious Diseases of the NationalInstitutes of Health (R01 AI111965). Y.-W.L. is the recipient of an Australian postgrad-uate award. J.L. is an Australian NHMRC senior research fellow. This study utilized theAustralian Phenomics Network Histopathology and Organ Pathology Service at theUniversity of Melbourne.

The content is solely the responsibility of the authors and does not necessarilyrepresent the official views of the National Institute of Allergy and Infectious Diseasesor the National Institutes of Health.

We thank Tien Nguyen for her assistance.

REFERENCES1. Arias CA, Murray BE. 2009. Antibiotic-resistant bugs in the 21st cen-

tury—a clinical super-challenge. N Engl J Med 360:439 – 443. https://doi.org/10.1056/NEJMp0804651.

2. Döring G, Conway S, Heijerman H, Hodson M, Høiby N, Smyth A, TouwD. 2000. Antibiotic therapy against Pseudomonas aeruginosa in cysticfibrosis: a European consensus. Eur Respir J 16:749 –767. https://doi.org/10.1034/j.1399-3003.2000.16d30.x.

3. Talbot GH, Bradley J, Edwards JE, Gilbert D, Scheld M, Bartlett JG.2006. Bad bugs need drugs: an update on the development pipelinefrom the Antimicrobial Availability Task Force of the Infectious Dis-eases Society of America. Clin Infect Dis 42:657– 668. https://doi.org/10.1086/499819.

4. Nation RL, Li J, Cars O, Couet W, Dudley MN, Kaye KS, Mouton JW,Paterson DL, Tam VH, Theuretzbacher U. 2015. Framework for optimi-sation of the clinical use of colistin and polymyxin B: the Prato polymyxinconsensus. Lancet Infect Dis 15:225–234. https://doi.org/10.1016/S1473-3099(14)70850-3.

5. Li J, Nation RL, Milne RW, Turnidge JD, Coulthard K. 2005. Evaluation of

colistin as an agent against multi-resistant Gram-negative bacteria. Int JAntimicrob Agents 25:11–25. https://doi.org/10.1016/j.ijantimicag.2004.10.001.

6. Yapa SW, Li J, Porter CJ, Nation RL, Patel K, McIntosh MP. 2013. Popu-lation pharmacokinetics of colistin methanesulfonate in rats: achievingsustained lung concentrations of colistin for targeting respiratory infec-tions. Antimicrob Agents Chemother 57:5087–5095. https://doi.org/10.1128/AAC.01127-13.

7. Yapa SW. 2013. PhD thesis. Monash University, Melbourne, Victoria,Australia.

8. Lu Q, Girardi C, Zhang M, Bouhemad B, Louchahi K, Petitjean O, WalletF, Becquemin M-H, Le Naour G, Marquette C-H. 2010. Nebulized andintravenous colistin in experimental pneumonia caused by Pseudomo-nas aeruginosa. Intensive Care Med 36:1147–1155. https://doi.org/10.1007/s00134-010-1879-4.

9. Marchand S, Bouchene S, de Monte M, Guilleminault L, Montharu J,Cabrera M, Grégoire N, Gobin P, Diot P, Couet W. 2015. Pharmacokineticsof colistin methansulphonate (CMS) and colistin after CMS nebulisation

Lin et al. Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 10

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 11: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

in baboon monkeys. Pharm Res 32:3403–3414. https://doi.org/10.1007/s11095-015-1716-0.

10. Gontijo AVL, Grégoire N, Lamarche I, Gobin P, Couet W, Marchand S.2014. Biopharmaceutical characterization of nebulized antimicrobialagents in rats: 2. Colistin. Antimicrob Agents Chemother 58:3950 –3956.https://doi.org/10.1128/AAC.02819-14.

11. Cheah S-E, Wang J, Turnidge JD, Li J, Nation RL. 2015. Newpharmacokinetic/pharmacodynamic studies of systemically adminis-tered colistin against Pseudomonas aeruginosa and Acinetobacter bau-mannii in mouse thigh and lung infection models: smaller response inlung infection. J Antimicrob Chemother 70:3291–3297. https://doi.org/10.1093/jac/dkv267.

12. Garonzik SM, Li J, Thamlikitkul V, Paterson DL, Shoham S, Jacob J, SilveiraFP, Forrest A, Nation RL. 2011. Population pharmacokinetics of colistinmethanesulfonate and formed colistin in critically ill patients from amulticenter study provide dosing suggestions for various categories ofpatients. Antimicrob Agents Chemother 55:3284 –3294. https://doi.org/10.1128/AAC.01733-10.

13. Hartzell JD, Neff R, Ake J, Howard R, Olson S, Paolino K, Vishnepolsky M,Weintrob A, Wortmann G. 2009. Nephrotoxicity associated with intrave-nous colistin (colistimethate sodium) treatment at a tertiary care medicalcenter. Clin Infect Dis 48:1724 –1728. https://doi.org/10.1086/599225.

14. Landersdorfer CB, Nguyen T-H, Lieu LT, Nguyen G, Bischof RJ, MeeusenEN, Li J, Nation RL, McIntosh MP. 2017. Substantial targeting advantageachieved by pulmonary administration of colistin methanesulfonate in alarge-animal model. Antimicrob Agents Chemother 61:e01934-16.https://doi.org/10.1128/AAC.01934-16.

15. Yapa SWS, Li J, Patel K, Wilson JW, Dooley MJ, George J, Clark D, PooleS, Williams E, Porter CJ, Nation RL, McIntosh MP. 2014. Pulmonary andsystemic pharmacokinetics of inhaled and intravenous colistin methane-sulfonate in cystic fibrosis patients: targeting advantage of inhalationaladministration. Antimicrob Agents Chemother 58:2570 –2579. https://doi.org/10.1128/AAC.01705-13.

16. Boisson M, Jacobs M, Grégoire N, Gobin P, Marchand S, Couet W, MimozO. 2014. Comparison of intrapulmonary and systemic pharmacokineticsof colistin methanesulfonate (CMS) and colistin after aerosol deliveryand intravenous administration of CMS in critically ill patients. Antimi-crob Agents Chemother 58:7331–7339. https://doi.org/10.1128/AAC.03510-14.

17. Lin Y-W, Zhou QT, Cheah S-E, Zhao J, Chen K, Wang J, Chan H-K, Li J.2017. Pharmacokinetics/pharmacodynamics of pulmonary delivery ofcolistin against Pseudomonas aeruginosa in a mouse lung infectionmodel. Antimicrob Agents Chemother 61:e02025-16. https://doi.org/10.1128/AAC.02025-16.

18. Touw D, Brimicombe R, Hodson M, Heijerman H, Bakker W. 1995. Inha-lation of antibiotics in cystic fibrosis. Eur Respir J 8:1594 –1604.

19. Mukhopadhyay S, Singh M, Cater J, Ogston S, Franklin M, Olver R. 1996.Nebulised antipseudomonal antibiotic therapy in cystic fibrosis: a meta-analysis of benefits and risks. Thorax 51:364 –368. https://doi.org/10.1136/thx.51.4.364.

20. Hodson M, Gallagher C, Govan J. 2002. A randomised clinical trial ofnebulised tobramycin or colistin in cystic fibrosis. Eur Respir J 20:658 – 664. https://doi.org/10.1183/09031936.02.00248102.

21. Pilcer G, Amighi K. 2010. Formulation strategy and use of excipients inpulmonary drug delivery. Int J Pharm 392:1–19. https://doi.org/10.1016/j.ijpharm.2010.03.017.

22. Zhou QT, Morton DA, Yu HH, Jacob J, Wang J, Li J, Chan HK. 2013.Colistin powders with high aerosolisation efficiency for respiratoryinfection: preparation and in vitro evaluation. J Pharm Sci 102:3736 –3747. https://doi.org/10.1002/jps.23685.

23. De Boer A, Le Brun P, Van der Woude H, Hagedoorn P, Heijerman H,Frijlink H. 2002. Dry powder inhalation of antibiotics in cystic fibrosistherapy, part 1: development of a powder formulation with colistinsulfate for a special test inhaler with an air classifier as de-agglomerationprinciple. Eur J Pharm Biopharm 54:17–24. https://doi.org/10.1016/S0939-6411(02)00043-7.

24. Le Brun P, De Boer A, Mannes G, de Fraîture DM, Brimicombe R, Touw D,Vinks A, Frijlink H, Heijerman H. 2002. Dry powder inhalation of antibi-otics in cystic fibrosis therapy: part 2: inhalation of a novel colistin drypowder formulation: a feasibility study in healthy volunteers and pa-tients. Eur J Pharm Biopharm 54:25–32. https://doi.org/10.1016/S0939-6411(02)00044-9.

25. Westerman EM, de Boer AH, Le Brun PP, Touw DJ, Frijlink HW, HeijermanHG. 2007. Dry powder inhalation of colistin sulphomethate in healthy

volunteers: a pilot study. Int J Pharm 335:41– 45. https://doi.org/10.1016/j.ijpharm.2006.11.021.

26. Westerman EM, De Boer AH, Le Brun PP, Touw DJ, Roldaan AC, FrijlinkHW, Heijerman HG. 2007. Dry powder inhalation of colistin in cysticfibrosis patients: a single dose pilot study. J Cyst Fibros 6:284 –292.https://doi.org/10.1016/j.jcf.2006.10.010.

27. Westerman E, Heijerman H, Frijlink H. 2007. Dry powder inhalation versuswet nebulisation delivery of antibiotics in cystic fibrosis patients. ExpertOpin Drug Deliv 4:91–94. https://doi.org/10.1517/17425247.4.2.91.

28. Reychler G, Keyeux A, Cremers C, Veriter C, Rodenstein DO, Liistro G.2004. Comparison of lung deposition in two types of nebulization:intrapulmonary percussive ventilation vs jet nebulization. Chest J 125:502–508. https://doi.org/10.1378/chest.125.2.502.

29. Abbott J, Dodd M, Bilton D, Webb A. 1994. Treatment compliance inadults with cystic fibrosis. Thorax 49:115–120. https://doi.org/10.1136/thx.49.2.115.

30. Burrows JA, Bunting JP, Masel PJ, Bell SC. 2002. Nebulised dornase alpha:adherence in adults with cystic fibrosis. J Cyst Fibros 1:255–259. https://doi.org/10.1016/S1569-1993(02)00095-4.

31. de Boer AH, Hagedoorn P, Westerman EM, Le Brun PP, Heijerman HG,Frijlink HW. 2006. Design and in vitro performance testing of multiple airclassifier technology in a new disposable inhaler concept (Twincer®) forhigh powder doses. Eur J Pharm Sci 28:171–178. https://doi.org/10.1016/j.ejps.2005.11.013.

32. Velkov T, Rahim NA, Zhou QT, Chan H-K, Li J. 2015. Inhaled anti-infectivechemotherapy for respiratory tract infections: successes, challenges andthe road ahead. Adv Drug Deliv Rev 85:65– 82. https://doi.org/10.1016/j.addr.2014.11.004.

33. Zhou QT, Tang P, Leung SSY, Chan JGY, Chan H-K. 2014. Emerginginhalation aerosol devices and strategies: Where are we headed? AdvDrug Deliv Rev 75:3–17. https://doi.org/10.1016/j.addr.2014.03.006.

34. VanDevanter DR, Geller DE. 2011. Tobramycin administered by the TOBI®Podhaler® for persons with cystic fibrosis: a review. Med Devices (Auckl)4:179 –188. https://doi.org/10.2147/MDER.S16360.

35. Schuster A, Haliburn C, Döring G, Goldman MH, Freedom Study Group.2013. Safety, efficacy and convenience of colistimethate sodium drypowder for inhalation (Colobreathe DPI) in patients with cystic fibrosis:a randomised study. Thorax 68:344 –350. https://doi.org/10.1136/thoraxjnl-2012-202059.

36. Marchand S, Gobin P, Brillault J, Baptista S, Adier C, Olivier J-C, Mimoz O,Couet W. 2010. Aerosol therapy with colistin methanesulfonate: a bio-pharmaceutical issue illustrated in rats. Antimicrob Agents Chemother54:3702–3707. https://doi.org/10.1128/AAC.00411-10.

37. Ratjen F, Rietschel E, Kasel D, Schwiertz R, Starke K, Beier H, VanKoningsbruggen S, Grasemann H. 2006. Pharmacokinetics of inhaledcolistin in patients with cystic fibrosis. J Antimicrob Chemother 57:306 –311. https://doi.org/10.1093/jac/dki461.

38. Athanassa ZE, Markantonis SL, Fousteri M-ZF, Myrianthefs PM, Bout-zouka EG, Tsakris A, Baltopoulos GJ. 2012. Pharmacokinetics of inhaledcolistimethate sodium (CMS) in mechanically ventilated critically ill pa-tients. Intensive Care Med 38:1779 –1786. https://doi.org/10.1007/s00134-012-2628-7.

39. Nakwan N, Lertpichaluk P, Chokephaibulkit K, Villani P, Regazzi M,Imberti R. 2015. Pulmonary and systemic pharmacokinetics of colistinfollowing a single dose of nebulized colistimethate in mechanicallyventilated neonates. Pediatr Infect Dis J 34:961–963. https://doi.org/10.1097/INF.0000000000000775.

40. Zhou QT, Leung SS, Tang P, Parumasivam T, Loh ZH, Chan HK. 2015.Inhaled formulations and pulmonary drug delivery systems for respira-tory infections. Adv Drug Deliv Rev 85:83–99. https://doi.org/10.1016/j.addr.2014.10.022.

41. Lin YW, Wong J, Qu L, Chan HK, Zhou QT. 2015. Powder production andparticle engineering for dry powder inhaler formulations. Curr Pharm Des21:3902–3916. https://doi.org/10.2174/1381612821666150820111134.

42. Jensen T, Pedersen SS, Garne S, Heilmann C, Høiby N, Koch C. 1987.Colistin inhalation therapy in cystic fibrosis patients with chronic Pseu-domonas aeruginosa lung infection. J Antimicrob Chemother 19:831– 838. https://doi.org/10.1093/jac/19.6.831.

43. Hansen C, Pressler T, Høiby N. 2008. Early aggressive eradication therapyfor intermittent Pseudomonas aeruginosa airway colonization in cysticfibrosis patients: 15 years experience. J Cyst Fibros 7:523–530. https://doi.org/10.1016/j.jcf.2008.06.009.

44. Chastre J, Fagon J-Y. 2002. Ventilator-associated pneumonia. Am J Respir

PK of Aerosolized Colistin Dry Powder Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 11

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from

Page 12: Pulmonary Pharmacokinetics of Colistin following ... · Pulmonary Pharmacokinetics of Colistin following Administration of Dry Powder Aerosols in Rats Yu-Wei Lin, aQi Tony Zhou,b

Crit Care Med 165:867–903. https://doi.org/10.1164/ajrccm.165.7.2105078.

45. Michalopoulos A, Kasiakou SK, Mastora Z, Rellos K, Kapaskelis AM,Falagas ME. 2005. Aerosolized colistin for the treatment of nosocomialpneumonia due to multidrug-resistant Gram-negative bacteria in pa-tients without cystic fibrosis. Crit Care 9:R53–R59. https://doi.org/10.1186/cc3020.

46. Li J, Milne RW, Nation RL, Turnidge JD, Smeaton TC, Coulthard K. 2003.Use of high-performance liquid chromatography to study the pharma-cokinetics of colistin sulfate in rats following intravenous administration.Antimicrob Agents Chemother 47:1766 –1770. https://doi.org/10.1128/AAC.47.5.1766-1770.2003.

47. Groneberg DA, Nickolaus M, Springer J, Döring F, Daniel H, Fischer A.2001. Localization of the peptide transporter PEPT2 in the lung: impli-cations for pulmonary oligopeptide uptake. Am J Pathol 158:707–714.https://doi.org/10.1016/S0002-9440(10)64013-8.

48. Groneberg D, Eynott P, Doring F, Thai D, Oates T, Barnes P, Chung K,Daniel H, Fischer A. 2002. Distribution and function of the peptidetransporter PEPT2 in normal and cystic fibrosis human lung. Thorax57:55– 60. https://doi.org/10.1136/thorax.57.1.55.

49. Kamal MA, Keep RF, Smith DE. 2008. Role and relevance of PEPT2 in drugdisposition, dynamics, and toxicity. Drug Metab Pharmacokinet 23:236 –242. https://doi.org/10.2133/dmpk.23.236.

50. Jin L, Zhou QT, Chan H-K, Larson IC, Pennington MW, Morales RA, BoydBJ, Norton RS, Nicolazzo JA. 2016. Pulmonary delivery of the Kv1.3-blocking peptide HsTX1 [R14A] for the treatment of autoimmunediseases. J Pharm Sci 105:650 – 656. https://doi.org/10.1016/j.xphs.2015.10.025.

51. Fernandes CA, Vanbever R. 2009. Preclinical models for pulmonary drugdelivery. Expert Opin Drug Deliv 6:1231–1245. https://doi.org/10.1517/17425240903241788.

52. Morello M, Krone CL, Dickerson S, Howerth E, Germishuizen WA, WongY-L, Edwards D, Bloom BR, Hondalus MK. 2009. Dry-powder pulmonaryinsufflation in the mouse for application to vaccine or drug studies.Tuberculosis (Edinb) 89:371–377. https://doi.org/10.1016/j.tube.2009.07.001.

53. Gales AC, Jones RN, Sader HS. 2011. Contemporary activity of colistinand polymyxin B against a worldwide collection of Gram-negativepathogens: results from the SENTRY antimicrobial surveillance program(2006-09). J Antimicrob Chemother 66:2070 –2074. https://doi.org/10.1093/jac/dkr239.

54. Huang JX, Blaskovich MA, Pelingon R, Ramu S, Kavanagh A, Elliott AG,Butler MS, Montgomery AB, Cooper MA. 2015. Mucin binding reducescolistin antimicrobial activity. Antimicrob Agents Chemother 59:5925–5931. https://doi.org/10.1128/AAC.00808-15.

55. Schwameis R, Erdogan-Yildirim Z, Manafi M, Zeitlinger M, Strommer S,Sauermann R. 2013. Effect of pulmonary surfactant on antimicrobialactivity in vitro. Antimicrob Agents Chemother 57:5151–5154. https://doi.org/10.1128/AAC.00778-13.

56. Manchandani P, Zhou J, Ledesma KR, Truong LD, Chow DS-L, Eriksen JL,Tam VH. 2016. Characterization of polymyxin B biodistribution anddisposition in an animal model. Antimicrob Agents Chemother 60:1029 –1034. https://doi.org/10.1128/AAC.02445-15.

57. Kunin CM, Bugg A. 1971. Binding of polymyxin antibiotics to tissues: the

major determinant of distribution and persistence in the body. J InfectDis 124:394 – 400. https://doi.org/10.1093/infdis/124.4.394.

58. Beringer P. 2001. The clinical use of colistin in patients with cysticfibrosis. Curr Opin Pulm Med 7:434 – 440. https://doi.org/10.1097/00063198-200111000-00013.

59. Kettler L, Sawyer S, Winefield H, Greville H. 2002. Determinants ofadherence in adults with cystic fibrosis. Thorax 57:459 – 464. https://doi.org/10.1136/thorax.57.5.459.

60. Sawicki GS, Sellers DE, Robinson WM. 2009. High treatment burden inadults with cystic fibrosis: challenges to disease self-management. J CystFibros 8:91–96. https://doi.org/10.1016/j.jcf.2008.09.007.

61. Conole D, Keating GM. 2014. Colistimethate sodium dry powder forinhalation: a review of its use in the treatment of chronic Pseudomonasaeruginosa infection in patients with cystic fibrosis. Drugs 74:377–387.https://doi.org/10.1007/s40265-014-0181-0.

62. Alothman GA, Ho B, Alsaadi MM, Ho SL, O’Drowsky L, Louca E, Coates AL.2005. Bronchial constriction and inhaled colistin in cystic fibrosis. ChestJ 127:522–529. https://doi.org/10.1378/chest.127.2.522.

63. Maddison J, Dodd M, Webb A. 1994. Nebulized colistin causes chesttightness in adults with cystic fibrosis. Respir Med 88:145–147. https://doi.org/10.1016/0954-6111(94)90028-0.

64. Cheah S-E, Bulitta JB, Li J, Nation RL. 2014. Development and validationof a liquid chromatography-mass spectrometry assay for polymyxin B inbacterial growth media. J Pharm Biomed Anal 92:177–182. https://doi.org/10.1016/j.jpba.2014.01.015.

65. Rennard S, Basset G, Lecossier D, O’Donnell K, Pinkston P, Martin P,Crystal R. 1986. Estimation of volume of epithelial lining fluid recoveredby lavage using urea as marker of dilution. J Appl Physiol 60:532–538.

66. Bulitta JB, Landersdorfer CB. 2011. Performance and robustness of theMonte Carlo importance sampling algorithm using parallelized S-ADAPTfor basic and complex mechanistic models. AAPS J 13:212–226. https://doi.org/10.1208/s12248-011-9258-9.

67. Beal SL. 2001. Ways to fit a PK model with some data below thequantification limit. J Pharmacokinet Pharmacodyn 28:481–504. https://doi.org/10.1023/A:1012299115260.

68. Landersdorfer CB, Kirkpatrick CM, Kinzig-Schippers M, Bulitta JB, Hol-zgrabe U, Drusano GL, Sörgel F. 2007. Population pharmacokinetics attwo dose levels and pharmacodynamic profiling of flucloxacillin. Anti-microb Agents Chemother 51:3290 –3297. https://doi.org/10.1128/AAC.01410-06.

69. Bulitta JB, Zhao P, Arnold RD, Kessler DR, Daifuku R, Pratt J, Luciano G,Hanauske A-R, Gelderblom H, Awada A. 2009. Mechanistic populationpharmacokinetics of total and unbound paclitaxel for a new nanodro-plet formulation versus Taxol in cancer patients. Cancer ChemotherPharmacol 63:1049 –1063. https://doi.org/10.1007/s00280-008-0827-2.

70. Tsuji BT, Okusanya OO, Bulitta JB, Forrest A, Bhavnani SM, Fernandez PB,Ambrose PG. 2011. Application of pharmacokinetic-pharmacodynamicmodeling and the justification of a novel fusidic acid dosing regimen:raising Lazarus from the dead. Clin Infect Dis 52:S513–S519. https://doi.org/10.1093/cid/cir166.

71. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. 2011. Prediction-corrected visual predictive checks for diagnosing nonlinear mixed-effects models. AAPS J 13:143–151. https://doi.org/10.1208/s12248-011-9255-z.

Lin et al. Antimicrobial Agents and Chemotherapy

November 2017 Volume 61 Issue 11 e00973-17 aac.asm.org 12

on April 7, 2020 by guest

http://aac.asm.org/

Dow

nloaded from