ArticlePDF Available

Population Pharmacokinetics of Polymyxin B: Population PK of Polymyxin B

Authors:
  • Astellas Pharma Global Development
  • NYU Langone Health

Abstract and Figures

Polymyxin B is used as a last treatment resort for multidrug-resistant Gram-negative bacterial infections. The objectives of this study were to examine the population pharmacokinetics of polymyxin B and investigate factor(s) influencing pharmacokinetic variability. Four serial blood samples each were collected from 35 adult patients at steady state. The concentrations of individual polymyxin B components were analyzed using a validated LCMS/MS assay and combined to derive total concentrations. A maximum likelihood expectation maximization approach was used to fit the data. Various demographic variables were investigated as potential covariates for clearance and volume of distribution (Vd ) using linear regression analysis. A 1-compartment model fit to the data satisfactorily (r2 = 0.96). The best-fit mean ± SD for clearance and Vd were 2.5 ± 1.1 L/h and 34.3 ± 16.4 L, respectively. Creatinine clearance was found to be a statistically significant covariate of clearance, but the magnitude was deemed clinically insignificant. This article is protected by copyright. All rights reserved.
Content may be subject to copyright.
Author Query Form
Journal: CPT
Article: 981
Dear Author,
During the copyediting of your manuscript the following queries arose.
Please refer to the query reference callout numbers in the page proofs and respond to each by marking the necessary com-
ments using the PDF annotation tools.
Please remember illegible or unclear comments and corrections may delay publication.
Many thanks for your assistance.
Query References Query Remarks?
AQ1 AUTHOR: Please confirm that given names (red) and surnames/family names (green) have been identified
correctly.
AQ0 AUTHOR: Please complete and return the publication charge form located on the landing page under the
Additional Forms section. We will be unable to post your corrected article online without this completed
form.
Funding Info Query Form
Please confirm that the funding sponsor list below was correctly extracted from your article: that it includes all funders and that the
text has been matched to the correct FundRef Registry organization names. If a name was not found in the FundRef registry, it may
be not the canonical name form or it may be a program name rather than an organization name, or it may be an organization not yet
included in FundRef Registry. If you know of another name form or a parent organization name for a “not found” item on this list
below, please share that information.
FundRef name FundRef Organization Name
Health Systems Research and Development Project (Faculty of
Medicine Siriraj Hospital, Thailand) [NOT FOUND IN FUNDREF REGISTRY]
Thai Health Promotion Fund Thai Health Promotion Foundation
Health Systems Research Institute (Thailand)
[NOT FOUND IN FUNDREF REGISTRY]
Government Pharmaceutical Organization (Thailand)
[NOT FOUND IN FUNDREF REGISTRY]
Singapore Ministry of Health Communicable Diseases Public
Health Research Grant [NOT FOUND IN FUNDREF REGISTRY]
Singapore National Medical Research Council Clinician Scien-
tist Award grant [NOT FOUND IN FUNDREF REGISTRY]
J_ID: CPT Customer A_ID: CPT981 Cadmus Art: CPT981 Ed. Ref. No.: 2017-0519 Date: 19-December-17 Stage: Page: 6
ID: nareshrao Time: 12:56 I Path: //chenas03/Cenpro/ApplicationFiles/Journals/Wiley/CPT#/Vol00000/170336/Comp/APPFile/JW-CPT#170336
Population Pharmacokinetics of Polymyxin B
AQ1 Pooja Manchandani
1
,Visanu Thamlikitkul
2
,Yanina Dubrovskaya
3
,Jessica T. Babic
4
,David C. Lye
5,6
,
Lawrence S. Lee
6
and Vincent H. Tam
1,7
Polymyxin B is used as a last treatment resort for multidrug-resistant Gram-negative bacterial infections. The objectives of
this study were to examine the population pharmacokinetics of polymyxin B and investigate factor(s) influencing pharmaco-
kinetic variability. Four serial blood samples each were collected from 35 adult patients at steady state. The concentrations
of individual polymyxin B components were analyzed using a validated liquid chromatography / tandem mass spectrometry
assay and combined to derive total concentrations. A maximum likelihood expectation maximization approach was used to
fit the data. Various demographic variables were investigated as potential covariates for clearance and volume of distribu-
tion (V
d
) using linear regression analysis. A one-compartment model fit to the data satisfactorily (r
2
50.96). The best-fit
mean 6SD for clearance and V
d
were 2.5 61.1 L/h and 34.3 616.4 L, respectively. Creatinine clearance was found to be
a statistically significant covariate of clearance, but the magnitude was deemed clinically insignificant.
Study Highlights
WHAT IS THE CURRENT KNOWLEDGE ON THE
TOPIC?
þDespite being the last treatment resort for resistant Gram-
negative bacterial infections, the clinical dosing of parenteral
polymyxin B is still not well established. This is partly attrib-
uted to the paucity of published reports on the clinical pharma-
cokinetics of polymyxin B. Furthermore, several studies in the
past have reported that polymyxin B is predominantly cleared
by nonrenal pathways. In addition to this, we have previously
demonstrated that polymyxin B exposures in patients with nor-
mal and impaired renal function after receiving standard dosing
of polymyxin B are comparable.
WHAT QUESTION DID THIS STUDY ADDRESS?
þThe study examined the population pharmacokinetics of
polymyxin B in patients with Gram-negative infections.
Moreover, the study investigated factors influencing the phar-
macokinetic variability of polymyxin B.
WHAT THIS STUDY ADDS TO OUR KNOWLEDGE
þWe showed that the total body clearance of polymyxin B
was not well correlated with the creatinine clearance of the
patients. In addition, the actual body weight of patients was
a poor predictor for the volume of distribution. Covariates
such as creatinine clearance and actual body weight of
patients might not be an accurate predictor of polymyxin B
pharmacokinetics.
HOW THIS MIGHT CHANGE CLINICAL PHARMA-
COLOGY OR TRANSLATIONAL SCIENCE
þThis population pharmacokinetic model would be a useful
tool in predicting the polymyxin B pharmacokinetic exposure
after standard clinical doses.
The increasing rate of antimicrobial resistance has posed a serious
threat to the clinical management of multidrug-resistant Gram-
negative bacterial infections.
1,2
This situation has led to the emer-
gence of parenteral polymyxins as a last-resort treatment option
against these challenging infections.
3–6
Both polymyxin B and
colistin (polymyxin E) are available and have been used clinically.
In view of its overall superior clinical pharmacological properties,
polymyxin B is expected to be more commonly used in the future.
7
Although polymyxin B is a potent antimicrobial agent, its clinical
utility is largely limited by its potential for nephrotoxicity.
8
Despite being available for clinical use for several decades, there is
still a great paucity of pharmacokinetic (PK) data guiding the opti-
mal dosing of polymyxin B in patients. Reports delineating the influ-
ence of various factors on the PK variability of polymyxin B in the
patients are scarce. As a result of these significant knowledge gaps,
the clinical dosing strategies of polymyxin B may not be fully opti-
mized. A thorough understanding of the clinical PKs of polymyxin B
is pivotal to maximize efficacy and minimize toxicity associated with
therapy. Therefore, the objectives of this study were to assess the PKs
of polymyxin B in a patient population and to investigate the factors
influencing PK variability. Combining relevant patient-specific sus-
ceptibility data, we anticipate that our findings would be a robust
tool in guiding the optimal polymyxin B treatment dosing strategy.
RESULTS
Patient demographics
Thirty-five patients (23 males, 4 Caucasians) were enrolled in the
study. The mean 6SD age, actual body weight, and creatinine
1
Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas, USA;
2
Faculty of Medicine Siriraj
Hospital, Mahidol University, Bangkok, Thailand;
3
Department of Pharmacy, New York University Langone Medical Center, New York, New York, USA;
4
Department of Pharmacy, Baylor St. Luke’s Medical Center, Houston, Texas, USA;
5
Institute of Infectious Diseases and Epidemiology, Tan Tock Seng
Hospital, Singapore;
6
Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore;
7
Department of Pharmacy
Practice and Translational Research, Houston, Texas, USA. Correspondence: Vincent H. Tam (vtam@uh.edu)
Received 25 July 2017; accepted 8 December 2017; advance online publication 00 Month 2017. doi:10.1002/cpt.981
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 00 NUMBER 00 | MONTH 2017 1
ARTICLES
J_ID: CPT Customer A_ID: CPT981 Cadmus Art: CPT981 Ed. Ref. No.: 2017-0519 Date: 19-December-17 Stage: Page: 1
ID: nareshrao Time: 12:56 I Path: //chenas03/Cenpro/ApplicationFiles/Journals/Wiley/CPT#/Vol00000/170336/Comp/APPFile/JW-CPT#170336
clearance of the patients included were 58.7 615.1 years, 57.7 6
15.6 kg, and 67 642 mL/min, respectively. Other demographic
details of the patients are summarized in TableT1 1.
Population PK modeling
A total of 139 data points were included in the analysis (Figure
S1, available online). The one-compartment model was chosen as
the best-fit model. The overall model fitting to the data was satis-
factory (r
2
50.96) and unbiased, as shown in FigureF1 1. The best-
fit PK parameter estimates and covariance matrix are shown in
Tables T22and T33. The mean elimination half-life was 10.1 h. Cre-
atinine clearance (but not age) was a statistically significant covar-
iate of clearance, but the magnitude was deemed clinically
insignificant (Figure S2). Volume of distribution (V
d
) was poorly
predicted by actual body weight (Figure S2). Furthermore, the
gender of the patients was not found to have a major effect on
the PK parameter estimates (data not shown). The PK profiles of
polymyxin B from selected dosing regimens were simulated based
on the best-fit model parameter estimates (Figure F22), and are
summarized in Tables T44and T55.
Correlation of drug exposure to outcome
Nephrotoxicity outcomes were available in 26 patients. In this
subcohort, the duration of therapy was 12.0 64.9 days. Overall,
the prevalence of nephrotoxicity was 26.9% (7 out of 26 patients;
Table 1 Demographic characteristics of patients
Characteristics
No. of patients 35
Male (%) 23 (65.7 %)
Age (years); mean 6SD (range) 58.7 615.1 (25–89)
Actual body weight (ABW) (kg); mean 6SD (range) 57.7 615.6 (36–112)
Total daily dose (mg); mean 6SD (range) 119.0 636.3 (65–240)
Dose/ABW (mg/kg); mean 6SD (range) 2.1 60.4 (1.3–2.8)
Baseline creatinine clearance (mL/min); mean 6SD (range) 66.8 642.4 (15–175)
Isolated micro-organisms
Acinetobacter baumannii 14
Pseudomonas aeruginosa 6
Klebsiella spp. 3
E. coli 2
Enterobacter spp. 1
Others 9
Infection site
Sputum 10
Blood 7
Abdomen 2
Urine 4
Others 12
Figure 1 Pred: predicted; Obs: Observed.
Table 2 Best-fit pharmacokinetic parameter estimates
Clearance (L/h) Volume of distribution (L)
Mean 2.5 34.3
Median 2.6 35.2
SD 1.1 16.4
% CV 43.8 47.8
% RES 9.0 11.3
SD, standard deviation; CV, coefficient of variation; RES, relative standard error.
J_ID: CPT Customer A_ID: CPT981 Cadmus Art: CPT981 Ed. Ref. No.: 2017-0519 Date: 19-December-17 Stage: Page: 2
ID: nareshrao Time: 12:56 I Path: //chenas03/Cenpro/ApplicationFiles/Journals/Wiley/CPT#/Vol00000/170336/Comp/APPFile/JW-CPT#170336
ARTICLES
2VOLUME 00 NUMBER 00 | MONTH 2017 | www.cpt-journal.com
two risk, five injury). The area under the curve (AUC) observed
in patients with and without nephrotoxicity were 52.3 6
14.8 mg.h/L and 45.1 617.3 mg.h/L, respectively (P50.31).
DISCUSSION
Parenteral polymyxin B has gained a revived interest with the
increasing prevalence of multidrug-resistance in Gram-negative
bacteria over the past few decades. Despite escalating clinical use,
the PKs of polymyxin B in humans is still not thoroughly
characterized.
Several studies have been conducted over last decade to bridge
the knowledge gap in our understanding of polymyxin B PKs.
Our research group published the first paper focusing on the pop-
ulation PKs of polymyxin B close to 10 years ago.
9
However,
only polymyxin B1 (the most abundant component in the poly-
myxin B USP mixture) was examined in a handful of patients
(n59) with relatively normal renal function. Another small
study (n58) provided additional insights that drug elimination
was predominantly via nonrenal clearance pathways.
10
More
recently, a larger population PK study examined 24 critically ill
patients with a range of renal function.
11
The authors reported
that total body clearance of polymyxin B was poorly predicted by
creatinine clearance.
To best of our knowledge, the study is the largest study con-
ducted to date, aimed at evaluating the population PKs of poly-
myxin B and identifying the patient factors influencing the PK
variability of the drug. In contrast to the previous studies men-
tioned above, the present study is multicentered, with the inclu-
sion of a wide range of subject ethnicity, renal function, and
standards of practice. The major components of commercially
available polymyxin B are polymyxin B1, B2, B3, and isoleucine
B1, which were reported to constitute 73.5%, 13.7%, 4.2%, and
8.6% of the mixture, respectively.
12
While individual polymyxin
B components were assayed individually, they were combined as
total polymyxin B concentrations for the purpose of assessing the
PKs. We have previously examined the relative concentration–
time courses of the major components of polymyxin B, and
found individual components of polymyxin B mixture exhibited
comparable PK profiles.
13
Similarly, no considerable difference in
in vitro potency was observed among the polymyxin B compo-
nents against several clinically important bacterial strains.
14
Our results showed that the mean polymyxin B clearance in
our diverse patient cohort was 2.5 L/h, which was not drastically
different from those reported previously (range 1.9–2.4 L/h).
9,11
In addition, the intersubject variability (% CV) observed was also
comparable to the recent study by Sandri et al. (43.8% vs.
32.4%).
11
However, the mean elimination half-life (10.1 h) was
shorter than those reported previously (range 11.9–13.6 h), but it
was not skewed predominantly by patients with augmented renal
function (i.e., creatinine clearance >140 mL/min) (data not
shown). Of note, observations were only made over one dosing
interval for each subject.
We evaluated both age and creatinine clearance as covariates of
total polymyxin B clearance, as we reasoned that the variables
Figure 2 Simulation profiles based on best-fit population PK model; total
daily dose of 100 mg administered at different dosing frequencies: Q24h,
Q12h and Q8h (a); different daily doses (100, 150, and 200 mg) adminis-
tered Q12h (b).
Table 3 Best-fit pharmacokinetic parameter covariance matrix
Clearance (L/h) Volume of distribution (L)
Clearance (L/h) 1.2
Volume of distribution (L) 12.8 269.8
Table 4 Simulation profiles based on best-fit pharmacokinetic
model when total daily dose of 100 mg of polymyxin B was
administered at different dosing frequencies
Concentration (mg/ml)
Polymyxin B dosing frequency
Q8h Q12h Q24h
Css trough 0.7 1.1 0.6
Css max 2.1 2.4 3.4
Css avg 1.3 1.6 1.6
Table 5 Simulation profiles based on best-fit pharmacokinetic
model when different daily dose was administered every 12 h
Concentration (mg/ml)
Total daily dose of polymyxin B
100 mg 150 mg 200 mg
Css trough 1.1 1.6 2.2
Css max 2.4 3.6 4.8
Css avg 1.6 2.5 3.3
J_ID: CPT Customer A_ID: CPT981 Cadmus Art: CPT981 Ed. Ref. No.: 2017-0519 Date: 19-December-17 Stage: Page: 3
ID: nareshrao Time: 12:56 I Path: //chenas03/Cenpro/ApplicationFiles/Journals/Wiley/CPT#/Vol00000/170336/Comp/APPFile/JW-CPT#170336
ARTICLES
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 00 NUMBER 00 | MONTH 2017 3
were likely correlated. Despite a low coefficient of determination
(r
2
<0.3), creatinine clearance (but not age) was found to be a
statistically significant covariate of clearance. Since the intersub-
ject variability of drug clearance in critically ill patients is
expected to range from 35–50%, a 2-fold change in AUC is com-
monly accepted as the threshold justification for dosing adjust-
ment. Using the best-fit regression equation (Figure S2), a 2-fold
change in AUC would be expected only when there is a greater
than 120 mL/min decrease in creatinine clearance. In clinical
practice, patients with such a level of deteriorating renal function
would likely have been placed on renal replacement therapy.
Consequently, the magnitude of association was deemed clinically
insignificant and the impact of these variables on clearance vari-
ability was not further pursued. The conclusion is in general
agreement with our previous observations excluding patients with
augmented renal function.
15
The patients in this study had a
wide range of body weight, but the best-fit volume of distribution
could not be predicted by any of the demographic variables
examined.
The overall nephrotoxicity prevalence rate observed in this
study was 26.9%, which was consistent with our previous findings
in the US of 21.1%.
8
A study conducted by Elias et al. correlated
the impact of polymyxin B dosage on the clinical outcomes (i.e.,
in-hospital mortality and severe renal impairment) of the
patients.
16
In contrast, we were unable to demonstrate a statisti-
cal difference in AUC among patients with and without nephro-
toxicity; a larger patient cohort might be necessary. Alternatively,
given the heterogeneous distribution of polymyxin B in different
organs,
17
drug concentration in renal tissues could be more infor-
mative than systemic drug exposure in predicting the likelihood/
onset of nephrotoxicity.
18
There are several limitations to this study. First, the dosing
guidelines were different for each clinical site and the dosing regi-
mens used for specific patients were not standardized. Second,
the renal function of the patients was not directly measured for
all patients. Third, the sample size was relatively small and might
have limited us in establishing a robust relationship between
polymyxin B exposures and nephrotoxicity. Finally, we did not
account for concomitant administration of other antibiotics and
their influence on the treatment efficacy/nephrotoxicity.
In conclusion, the PKs of polymyxin B in a patient population
were characterized. In conjunction with relevant patient-specific
data, we anticipate that our model could be extended as a tool to
predict the PK exposure of polymyxin B in patients.
METHODS
Study design and sites
This study was a prospective, multicenter, observational study conducted
at four clinical sites: Siriraj Hospital (a 2,300-bed academic tertiary care
hospital) in Bangkok, Thailand; Tan Tock Seng Hospital (a 1,400-bed
acute-care general hospital) in Singapore; New York University Langone
Medical Center (a 800-bed academic medical center) in New York, NY;
and Baylor St. Luke’s Medical Center (a 850-bed teaching hospital) in
Houston, TX. Institutional Review Board (IRB) approval at each clinical
site and the University of Houston was obtained prior to the initiation
of this study. Written informed consent was obtained from each patient
(or their legal representative) prior to study enrollment.
Inclusion/exclusion criteria for patient enrollment
Adult patients (age 18 years) who were given at least 48 h of intrave-
nous polymyxin B (USP) daily for suspected/documented Gram-
negative bacterial infections were included in the study. Patients on any
form of renal replacement therapy or with fluctuating renal function
(increase or decrease in serum creatinine of more than 50% from the first
day of polymyxin therapy) were excluded.
Drug administration
Polymyxin B dosing regimens (daily dose, dosing interval, duration of
therapy, and duration of intravenous administration) at different clinical
sites were at the discretion of their attending medical teams. Polymyxin
B was administered as an intermittent intravenous infusion over 60–180
minutes, and every 12 h to 24 h.
Data collection
Patient data collected included demographics (e.g., age, ethnicity, and
gender) and pertinent laboratory findings (e.g., isolated micro-organism,
infection site, and serum creatinine). Creatinine clearance was estimated
from serum creatinine using the Cockcroft–Gault equation or urine col-
lection (by each clinical site). The prevalence of nephrotoxicity, as
defined according to the RIFLE (risk, injury, failure, loss, endstage kid-
ney disease) criteria
19
was tracked in selected patients for the duration of
polymyxin B therapy.
PK sampling schedule and polymyxin B assay
Four serial blood samples were collected from each patient at steady state
over the fourth or greater dosing interval (e.g., immediately prior to dos-
ing, 1–2 h, 8–12 after the end of drug infusion, and prior to the next
dose). Plasma samples were obtained by centrifugation within 30 min of
blood collection and were stored at –808C until analysis. Major compo-
nents of polymyxin B (e.g., polymyxin B1, polymyxin B2, polymyxin B3,
and isoleucine polymyxin B1) were quantified using a validated UP liq-
uid chromatography / tandem mass spectrometry (LC-MS/MS) method
with few modifications: Acquity UPLC HSS C
18
column (50 32.1 mm
internal diameter, 1.7 lm) from Waters (Milford, MA); mobile phase A,
0.1% formic acid in water; and mobile phase B, 0.1% formic acid in ace-
tonitrile. The samples were analyzed as described previously.
20
Population PK modeling and statistical analysis
The plasma concentration of each component (polymyxin B1, B2, B3,
and isoleucine B1) was quantified individually, and the concentrations
were summed to derive the total polymyxin B concentrations for esti-
mating the PK parameters. A maximum likelihood expectation maximi-
zation approach (MLEM) using one- and two-compartment models
with log-normal distributions was used to fit the PK profiles.
21–23
A
change of <0.001% in the likelihood function was set as the convergence
criterion for each structural model. The structural models were discrimi-
nated using the log-likelihood ratio test, adjusted for the difference in
the degrees of freedom. All PK modeling was performed using ADAPT
5 (University of Southern California, Los Angeles, CA).
The best-fit population PK parameters, clearance (in liters per hour)
and volume of distribution (V
d
, in liters), are reported as mean 6SD.
Using these best-fit parameter estimates and different dosing regimens,
the corresponding C
max
and C
min
were determined. Also, the area under
the concentration–time profile (AUC) of each subject was determined
by patient-specific daily dose/clearance. Various demographic variables
such as age, gender, ethnicity, actual body weight, and creatinine clear-
ance were investigated as potential covariates for the best-fit clearance
and V
d
. Continuous variables were compared using Student’s t-test. Cat-
egorical variables were compared using Fisher’s exact test. The influence
of potential covariates on the variability of PK parameters was estimated
by linear regression analysis. Pvalues of 0.05 or less were considered sig-
nificant. All the statistical analysis was performed using SYSTAT v. 12.0
(SYSTAT Software, Chicago, IL).
J_ID: CPT Customer A_ID: CPT981 Cadmus Art: CPT981 Ed. Ref. No.: 2017-0519 Date: 19-December-17 Stage: Page: 4
ID: nareshrao Time: 12:56 I Path: //chenas03/Cenpro/ApplicationFiles/Journals/Wiley/CPT#/Vol00000/170336/Comp/APPFile/JW-CPT#170336
ARTICLES
4VOLUME 00 NUMBER 00 | MONTH 2017 | www.cpt-journal.com
AUTHOR CONTRIBUTIONS
P.M. and V.H.T. wrote the article; V.H.T. designed the research; V.T.,
Y.D., J.T.B., and D.C.L. performed the research; P.M. and V.H.T. analyzed
the data; P.M. and L.S.L. contributed new reagents/analytical tools.
FUNDING
The study was supported in part by the Health Systems Research and
Development Project (Faculty of Medicine Siriraj Hospital, Thailand), the
Thai Health Promotion Fund, the Health Systems Research Institute
(Thailand), Government Pharmaceutical Organization (Thailand), the Sin-
gapore Ministry of Health Communicable Diseases Public Health
Research Grant (CDPHRG/12NOV015) and the Singapore National Medi-
cal Research Council Clinician Scientist Award grant (NMRC/CSAINV/
0005/2016). The funders had no role in study design, data collection
and interpretation, or the decision to submit the work for publication.
Additional Supporting Information may be found in the online version of
this article.
CONFLICT OF INTEREST
The authors report no conflicts of interest.
V
C2017 American Society for Clinical Pharmacology and Therapeutics
1. Karaiskos, I. & Giamarellou, H. Multidrug-resistant and extensively
drug-resistant Gram-negative pathogens: current and emerging
therapeutic approaches. Expert Opin. Pharmacother. 15, 1351–1370
(2014).
2. Vergidis, P.I. & Falagas, M.E. Multidrug-resistant Gram-negative
bacterial infections: the emerging threat and potential novel
treatment options. Curr. Opin. Investig. Drugs 9, 176–183 (2008).
3. Huttner, B., Jones, M., Rubin, M.A., Neuhauser, M.M., Gundlapalli, A.
& Samore, M. Drugs of last resort? The use of polymyxins and
tigecycline at US Veterans Affairs medical centers, 2005-2010. PLoS
One 7, e36649 (2012).
4. Arnold, T.M., Forrest, G.N. & Messmer, K.J. Polymyxin antibiotics for
gram-negative infections. Am. J. Health Syst. Pharm. 64, 819–826
(2007).
5. Pastewski, A.A. et al. Parenteral polymyxin B use in patients with
multidrug-resistant gram-negative bacteremia and urinary tract
infections: a retrospective case series. Ann. Pharmacother. 42,
1177–1187 (2008).
6. Delgado-Valverde, M., Sojo-Dorado, J., Pascual, A. & Rodriguez-Bano,
J. Clinical management of infections caused by multidrug-resistant
Enterobacteriaceae. Ther. Adv. Infect. Dis. 1, 49–69 (2013).
7. Nation, R.L., Velkov, T. & Li, J. Colistin and polymyxin B: peas in a
pod, or chalk and cheese? Clin. Infect. Dis. 59, 88–94 (2014).
8. Phe, K. et al. In vitro assessment and multicenter cohort study of
comparative nephrotoxicity rates associated with colistimethate
versus polymyxin b therapy. Antimicrob. Agents Chemother. 58,
2740–2746 (2014).
9. Kwa, A.L. et al. Pharmacokinetics of polymyxin B1 in patients with
multidrug-resistant Gram-negative bacterial infections. Diagn.
Microbiol. Infect. Dis. 60, 163–167 (2008).
10. Zavascki, A.P. et al. Pharmacokinetics of intravenous polymyxin B in
critically ill patients. Clin. Infect. Dis. 47, 1298–1304 (2008).
11. Sandri, A.M. et al. Population pharmacokinetics of intravenous
polymyxin B in critically ill patients: implications for selection of
dosage regimens. Clin. Infect. Dis. 57, 524–531 (2013).
12. He, J., Gao, S., Hu, M., Chow, D.S. & Tam, V.H. A validated ultra-
performance liquid chromatography-tandem mass spectrometry
method for the quantification of polymyxin B in mouse serum and
epithelial lining fluid: application to pharmacokinetic studies. J.
Antimicrob. Chemother. 68, 1104–1110 (2013).
13. Manchandani, P., Dubrovskaya, Y., Gao, S. & Tam, V.H. Comparative
Pharmacokinetic Profiling of Different Polymyxin B Components.
Antimicrob. Agents Chemother. 60, 6980–6982 (2016).
14. Tam, V.H., Cao, H., Ledesma, K.R. & Hu, M. In vitro potency of
various polymyxin B components. Antimicrob. Agents Chemother. 55,
4490–4491 (2011).
15. Thamlikitkul, V. et al. Dosing and Pharmacokinetics of Polymyxin B in
Patients with Renal Insufficiency. Antimicrob. Agents Chemother. 61,
e01337–01316 (2017).
16. Elias, L.S., Konzen, D., Krebs, J.M. & Zavascki, A.P. The impact of
polymyxin B dosage on in-hospital mortality of patients treated with
this antibiotic. J. Antimicrob. Chemother. 65, 2231–2237 (2010).
17. Manchandani, P. et al. Characterization of polymyxin B biodistribution
and disposition in an animal model. Antimicrob. Agents Chemother.
60, 1029–1034 (2016).
18. Manchandani, P., Zhou, J., Babic, J.T., Ledesma, K.R., Truong, L.D. &
Tam, V.H. Role of renal drug exposure in polymyxin B-induced
nephrotoxicity. Antimicrob. Agents Chemother. 61, e02391–02316
(2017).
19. Bellomo, R., Ronco, C., Kellum, J.A., Mehta, R.L., Palevsky, P. &
Acute Dialysis Quality Initiative, w. Acute renal failure — definition,
outcome measures, animal models, fluid therapy and information
technology needs: the Second International Consensus Conference
of the Acute Dialysis Quality Initiative (ADQI) Group. Crit. Care 8,
R204–212 (2004).
20. Hee, K.H., Leaw, Y.K.J., Ong, J.L. & Lee, L.S. Development and
validation of liquid chromatography tandem mass spectrometry
method quantitative determination of polymyxin B1, polymyxin B2,
polymyxin B3 and isoleucine-polymyxin B1 in human plasma and its
application in clinical studies. J. Pharm. Biomed. Anal. 140, 91–97
(2017).
21. Colucci, P., Grenier, J., Yue, C.S., Turgeon, J. & Ducharme, M.P.
Performance of different population pharmacokinetic algorithms.
Ther. Drug Monit. 33, 583–591 (2011).
22. Plan, E.L., Maloney, A., Troconiz, I.F. & Karlsson, M.O. Performance
in population models for count data, part I: maximum likelihood
approximations. J. Pharmacokinet. Pharmacodyn. 36, 353–366
(2009).
23. Savic, R. & Lavielle, M. Performance in population models for count
data, part II: a new SAEM algorithm. J. Pharmacokinet. Pharmacodyn.
36, 367–379 (2009).
J_ID: CPT Customer A_ID: CPT981 Cadmus Art: CPT981 Ed. Ref. No.: 2017-0519 Date: 19-December-17 Stage: Page: 5
ID: nareshrao Time: 12:56 I Path: //chenas03/Cenpro/ApplicationFiles/Journals/Wiley/CPT#/Vol00000/170336/Comp/APPFile/JW-CPT#170336
ARTICLES
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 00 NUMBER 00 | MONTH 2017 5
... A comprehensive understanding of the significant variability in the pharmacokinetic profile of PMB assists in the development of individualization dosing strategies [4,5,13,14]. Accordingly, several previous studies have been performed to characterize PMB pharmacokinetics, including the development of the PopPK models to quantify and explain inter-individual variability (IIV) for dose individualization in patients [14][15][16][17][18][19][20][21][22][23][24][25][26][27][28]. These studies have identified various covariates (e.g., total body weight [TBW], creatinine clearance [CrCL]) that influence PMB clearance (CL) and volume of distribution (V), however, the included covariates only partially explain the observed IIV and interoccasion variability (IOV) [16,19,23,24,27]. ...
... Although several PMB PopPK models have been published to assess individualized dose adjustment [17,20,24,25], these PopPK models were established in specific populations. Additionally, considering the variation among different centers, evaluating the accuracy and applicability of these models is essential [29][30][31][32]. ...
... This population consisted of four different groups: patients with liver dysfunction, kidney dysfunction, liver and kidney dysfunction, and normal liver and kidney function. Additionally, liver function, as a very important factor affecting drug metabolism, has not been extensively discussed in the published literature [15][16][17][18][19][20][21][22][23][24][25][26][27][28]. To the best of our knowledge, this ...
Article
Full-text available
Polymyxin B (PMB) is considered a last-line treatment for multidrug-resistant (MDR) gram-negative bacterial infections. Model-informed precision dosing with population pharmacokinetics (PopPK) models could help to individualize PMB dosing regimens and improve therapy. However, the external prediction ability of the established PopPK models has not been fully elaborated. This study aimed to systemically evaluate eleven PMB PopPK models from ten published literature based on a new independent population, which was divided into four different populations, patients with liver dysfunction, kidney dysfunction, liver and kidney dysfunction, and normal liver and kidney function. The whole data set consisted of 146 patients with 391 PMB concentrations. The prediction- and simulation-based diagnostics and Bayesian forecasting were conducted to evaluate model predictability. In the overall evaluation process, none of the models exhibited satisfactory predictive ability in both prediction- and simulation-based diagnostic simultaneously. However, the evaluation of the models in the subgroup of patients with normal liver and kidney function revealed improved predictive performance compared to those with liver and/or kidney dysfunction. Bayesian forecasting demonstrated enhanced predictability with the incorporation of two to three prior observations. The external evaluation highlighted a lack of consistency between the prediction results of published models and the external validation dataset. Nonetheless, Bayesian forecasting holds promise in improving the predictive performance of the models, and feedback from therapeutic drug monitoring is crucial in optimizing individual dosing regimens.
... The current international guideline recommends that the dose of PMB should be calculated based on the patient's weight, irrespective of age (Tsuji et al., 2019). Several population pharmacokinetics (PK) studies have reported that PMB clearance is independent of age in critically ill patients or patients with cystic fibrosis (Sandri et al., 2013;Manchandani et al., 2018;Wang et al., 2020), whereas one study has found that age is significantly related with the volume of distribution of PMB (Liang et al., 2023). Whether elderly patients have higher PMB exposure than young critically ill patients and different clearance and volume of distribution from young patients given the standard dose has remained unknown until now. ...
... To our knowledge, this is the first study to date comparing PMB exposure and other individual PK parameters in critically ill patients of different ages given the standard dosing of PMB. Age was not shown to be correlated with clearance and volume of distribution of PMB in critically ill patients in this study, consistent with many previously published studies (Sandri et al., 2013;Manchandani et al., 2018;Wang et al., 2020;Wang et al., 2021a;Yu et al., 2021;Surovoy et al., 2022;Yang et al., 2022). However, we did find that the elderly patients had longer half-life time than the young critically ill patients and age was positively correlated with half-life time of PMB. ...
... However, the relationship between TBW and clearance and volume of distribution of PMB was not certain. In contrast to our study, many PK studies did not find TBW as a covariate of the PMB clearance and volume of distribution Kubin et al., 2018;Manchandani et al., 2018;Wang et al., 2020;Wang et al., 2021a;Wang et al., 2021b;Li et al., 2021;Yu et al., 2021), and weight-based dosage regimens are being challenged based on these findings (Liu et al., 2023). Therefore, such discrepancy could be caused by several factors, Such as the difference in sample size, with a range of 9-70 (Sandri et al., 2013;Kubin et al., 2018;Manchandani et al., 2018;Miglis et al., 2018;Wang et al., 2020;Wang et al., 2021a;Wang et al., 2021b;Crass et al., 2021;Li et al., 2021;Yu et al., 2021), different compartment models, and different dosing regimens used in various studies. ...
Article
Full-text available
Introduction: Elderly patients are more prone to develop acute kidney injury during infections and polymyxin B (PMB)-associated nephrotoxicity than young patients. The differential response to PMB between the elderly and young critically ill patients is unknown. We aimed to assess PMB exposure in elderly patients compared with young critically ill patients, and to determine the covariates of PMB pharmacokinetics in critically ill patients. Methods: Seventeen elderly patients (age ≥ 65 years) and six young critically ill patients (age < 65 years) were enrolled. Six to eight blood samples were collected during the 12 h intervals after at least six doses of intravenous PMB in each patient. PMB plasma concentrations were quantified by high-performance liquid chromatography-tandem mass spectrometry. The primary outcome was PMB exposure as assessed by the area under the concentration-time curve over 24 h at steady state (AUCss, 0–24 h). Results and Discussion: The elderly group had lower total body weight (TBW) and higher Charlson comorbidity scores than young group. Neither AUCss, 0–24 h nor normalized AUCss, 0–24 h (adjusting AUC for the daily dose in mg/kg of TBW) was significantly different between the elderly group and young group. The half-life time was longer in the elderly patients than in young patients (11.21 vs 6.56 h respectively, p = 0.003). Age and TBW were the covariates of half-life time (r = 0.415, p = 0.049 and r = −0.489, p = 0.018, respectively). TBW was the covariate of clearance (r = 0.527, p = 0.010) and AUCss, 0–24 h (r = −0.414, p = 0.049). Patients with AUCss, 0–24 h ≥ 100 mg·h/L had higher baseline serum creatinine levels and lower TBW than patients with AUCss, 0–24 h < 50 mg·h/L or patients with AUCss, 0–24 h 50–100 mg·h/L. The PMB exposures were comparable in elderly and young critically ill patients. High baseline serum creatinine levels and low TBW was associated with PMB overdose. Trial registration: ChiCTR2300073896 retrospectively registered on 25 July 2023.
... Based on the validation group of 14 patients with severe infection, Luo et al. suggested that the final model performed superior for Css,max and Css,1/2t, but relatively inferior for Css,min [17]. Based on a data set of 20 Chinese (92 samples), Li et al.'s external validation results of the above 8 models revealed that only the models by Manchandani et al. [29] and Kubin et al. [24] were better than the others, with F20 > 40% and F30 > 50% in prediction-based diagnostics [30]. The models by Miglis et al. [23], Manchandani et al. [29], and Kubin et al. [24] demonstrated that the observed concentration was captured in the range of the CIs in simulation-based diagnostics, suggesting more favorable predictive power. ...
... Based on a data set of 20 Chinese (92 samples), Li et al.'s external validation results of the above 8 models revealed that only the models by Manchandani et al. [29] and Kubin et al. [24] were better than the others, with F20 > 40% and F30 > 50% in prediction-based diagnostics [30]. The models by Miglis et al. [23], Manchandani et al. [29], and Kubin et al. [24] demonstrated that the observed concentration was captured in the range of the CIs in simulation-based diagnostics, suggesting more favorable predictive power. While not all published models performed satisfactorily in simulation and prediction-based diagnostics overall, Bayesian forecasting enhanced predictability with priors [30]. ...
Article
Full-text available
Background and objectives Despite being clinically utilized for the treatment of infections, the limited therapeutic range of polymyxin B (PMB), along with considerable interpatient variability in its pharmacokinetics and frequent occurrence of acute kidney injury, has significantly hindered its widespread utilization. Recent research on the population pharmacokinetics of PMB has provided valuable insights. This study aims to review relevant literature to establish a theoretical foundation for individualized clinical management. Methods Follow PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, Pop-PK studies of PMB were searched in PubMed and EMBASE database systems from the inception of the database until March 2023. Result To date, a total of 22 population-based studies have been conducted, encompassing 756 subjects across six different countries. The recruited population in these studies consisted of critically infected individuals with multidrug-resistant bacteria, patients with varying renal functions, those with cystic fibrosis, kidney or lung transplant recipients, patients undergoing extracorporeal membrane oxygenation (ECMO) or continuous renal replacement therapy (CRRT), as well as individuals with obesity or pediatric populations. Among these studies, seven employed a one-compartmental model, with the range of typical clearance (CL) and volume (Vc) being 1.18–2.5L /h and 12.09–47.2 L, respectively. Fifteen studies employed a two-compartmental model, with the ranges of the clearance (CL) and volume of the central compartment (Vc), the volume of the peripheral compartment (Vp), and the intercompartment clearance (Q) were 1.27–8.65 L/h, 5.47–38.6 L, 4.52–174.69 L, and 1.34–24.3 L/h, respectively. Primary covariates identified in these studies included creatinine clearance and body weight, while other covariates considered were CRRT, albumin, age, and SOFA scores. Internal evaluation was conducted in 19 studies, with only one study being externally validated using an independent external dataset. Conclusion We conclude that small sample sizes, lack of multicentre collaboration, and patient homogeneity are the primary reasons for the discrepancies in the results of the current studies. In addition, most of the studies limited in the internal evaluation, which confined the implementation of model-informed precision dosing strategies.
... Polymyxins are lipopeptide antibiotics synthesized by a nonribosomal peptide synthase system, which was firstly isolated from Paenibacillus polymyxa (Storm et al. 1977;Martin et al. 2003). There are various types of polymyxins, among which polymyxin B and polymyxin E have effective inhibitory effects on multidrugresistant bacteria (Poirel et al. 2017;Manchandani et al. 2018;Nakwan et al. 2019). Unfortunately, the increased and inappropriate use of polymyxins has led inexorably to the worldwide emergence of polymyxin-resistant bacteria in both veterinary clinics and human medical clinics. ...
Article
Full-text available
Polymyxins are cationic peptide antibiotics and regarded as the “final line of defense” against multidrug-resistant bacterial infections. Meanwhile, some polymyxin-resistant strains and the corresponding resistance mechanisms have also been reported. However, the response of the polymyxin-producing strain Paenibacillus polymyxa to polymyxin stress remains unclear. The purpose of this study was to investigate the stress response of gram-positive P. polymyxa SC2 to polymyxin B and to identify functional genes involved in the stress response process. Polymyxin B treatment upregulated the expression of genes related to basal metabolism, transcriptional regulation, transport, and flagella formation and increased intracellular ROS levels, flagellar motility, and biofilm formation in P. polymyxa SC2. Adding magnesium, calcium, and iron alleviated the stress of polymyxin B on P. polymyxa SC2, furthermore, magnesium and calcium could improve the resistance of P. polymyxa SC2 to polymyxin B by promoting biofilm formation. Meanwhile, functional identification of differentially expressed genes indicated that an ABC superfamily transporter YwjA was involved in the stress response to polymyxin B of P. polymyxa SC2. This study provides an important reference for improving the resistance of P. polymyxa to polymyxins and increasing the yield of polymyxins. Key points • Phenotypic responses of P. polymyxa to polymyxin B was performed and indicated by RNA-seq • Forming biofilm was a key strategy of P. polymyxa to alleviate polymyxin stress • ABC transporter YwjA was involved in the stress resistance of P. polymyxa to polymyxin B
... Comparing AKI incidence, c P < 0.05 in the effective treatment group versus the ineffective treatment group resulting in cell swelling and cell lysis [16]. Mohammad et al. [17] detected that the concentration of polymyxin B in human renal tubular cells is approximately 4760 times higher than the extracellular concentration, indicating that the reabsorption of polymyxin B in renal tubules leads to abnormal accumulation in cells and direct damage to tubular cells. ...
Article
Full-text available
Background To study the efficacy and nephrotoxicity of polymyxin B in the treatment of elderly patients with carbapenem-resistant organism (CRO) infection. Methods The clinical and microbiological data of patients with CRO-infected sepsis treated with polymyxin B were retrospectively analyzed. The effective rate, bacterial clearance, incidence and recovery rate of acute renal injury (AKI) and prognosis-related indicators in AKI at different stages were compared. Results The effective rate of 215 elderly patients with CRO infection treated with polymyxin was 50.7%. The total bacterial clearance rate was 44.2%, the total incidence of AKI was 37.2%, the recovery rate of AKI was 35%, and the incidence range of polymyxin B-related AKI was 10.2–37.2%. Logistic multivariate regression analysis showed that the predictors of AKI in elderly patients were high APACHE II score, long duration of polymyxin, chronic renal insufficiency and ineffective outcome; the ROC curve showed that the cutoff value for predicting AKI was a serum creatinine concentration of 73 mmol/L before polymyxin B use, and the AUC was 0.931. Conclusions Rational use of polymyxin B is safe and effective in elderly patients with CRO infection, and its effective outcome can improve the recovery rate of AKI.
Article
Background: Polymyxins have been regarded as last-line treatment for multidrug-resistant gram-negative bacterial infections. Nonetheless, concerns regarding toxicity persist. This study aimed to explore and compare potential adverse events (AEs) between colistin and polymyxin B (PMB). Methods: Outpatient antibiotic use associated with acute upper respiratory infections in China: a nationwide cross-sectional study Polymyxins-related AEs were retrieved from the U.S. Food and Drug Administration Adverse Event Reporting System between 2004 and 2022. Potential signals were estimated by the reporting odds ratio (ROR), and subgroup analyses were preformed to adjust for potential factors in AEs with significant disproportionality. Results: Analysis of 3,915 records involving 718 patients revealed a higher disproportionality of renal and urinary disorders (ROR 1.62, 95% CI 1.01-2.59) and acute kidney injury (ROR 1.75, 95% CI 1.07-2.87) with colistin treatment. Conversely, colistin exhibited a lower risk for neurotoxicity (ROR 0.47, 95% CI 0.30-0.73). Seven cases of skin hyperpigmentation were reported with PMB, whereas none were reported with colistin. Over 80% of cases involving polymyxin-related AEs occurred during the first two weeks of therapies, with a median onset time of 4.5 days. Conclusions: Outpatient antibiotic use associated with acute upper respiratory infections in China: a nationwide cross-sectional study Patients received colistin displayed a higher potential risk of nephrotoxicity but a lower risk of neurotoxicity. Clinicians should be vigilant in monitoring the AEs of hyperpigmentation disorders induced by PMB.
Article
Full-text available
Aims Polymyxin B (PMB) is widely used to treat infections caused by multidrug‐resistant Gram‐negative pathogens. Currently, the pharmacokinetic data of PMB in patients with liver dysfunction are limited. This study aimed to develop a population pharmacokinetic (PopPK) model of PMB in patients with liver dysfunction and identify the factors affecting PMB pharmacokinetics. Methods We conducted a retrospective pharmacokinetic study involving 136 adults with different levels of liver function. Nonlinear mixed effects modelling was used to develop a PopPK model of PMB. Monte Carlo simulation was used to design PMB dosage schedules across various liver and renal functions. Results PMB pharmacokinetic analyses included 401 steady‐state concentrations in 136 adult patients. A one‐compartment pharmacokinetic model with first‐order absorption and elimination was used to describe the data. The typical population value of PMB clearance was 2.43 L/h and the volume of distribution was 23.11 L. This study revealed that creatinine clearance (CrCL) and Child–Pugh class were significantly associated with PMB pharmacokinetic parameters; however, clinically relevant variations of dose‐normalized drug exposure were not significant. For patients with a minimum inhibitory concentration of ≤0.5 mg/L, the appropriate dose was 40–75 mg/12‐h. When the dose exceeded 100 mg/12‐h, the risk of nephrotoxicity increased significantly. Conclusions This study provided PMB pharmacokinetic information for patients with liver dysfunction. Patients with renal and liver dysfunctions may not require an initial dose adjustment. Rather than PopPK‐guided dose adjustment, therapeutic drug monitoring of PMB plays a more direct role in optimizing dosing regimens based on its therapeutic window.
Article
Full-text available
Despite dose-limiting nephrotoxic potentials, polymyxin B has re-emerged as the last line of therapy against multidrug-resistant Gram-negative bacterial infections. However, the handling of polymyxin B by the kidneys is still not thoroughly understood. The objectives of this study were to evaluate the impact of renal polymyxin B exposure on nephrotoxicity and to explore the role of megalin in renal drug accumulation. Sprague-Dawley rats (225-250 g) were divided into 3 dosing groups, and polymyxin B was administered (5 mg/kg, 10 mg/kg and 20 mg/kg) subcutaneously once daily. The onset of nephrotoxicity over 7 days and renal drug concentrations 24h after the first dose were assessed. The effects of sodium maleate (400 mg/kg intraperitoneally) on megalin homeostasis were evaluated by determining urinary megalin concentration and electron microscopic study of renal tissue. The serum/renal pharmacokinetics of polymyxin B were assessed in megalin-shedding rats. The onset of nephrotoxicity was correlated to the daily dose of polymyxin B. Renal polymyxin B concentrations were found to be 3.6 ± 0.4 μg/g, 9.9 ± 1.5 μg/g and 21.7 ± 4.8 μg/g in the 5 mg/kg, 10 mg/kg and 20 mg/kg dosing groups, respectively. In megalin-shedding rats, the serum pharmacokinetics of polymyxin B remained unchanged, but the renal exposure was attenuated by 40 % as compared to control rats. Onset of polymyxin B-induced nephrotoxicity is correlated to the renal drug exposure. In addition, megalin appears to play a pivotal role in the renal accumulation of polymyxin B, which might contribute to nephrotoxicity.
Article
Full-text available
Polymyxin B remains the last-line treatment option for multidrug-resistant Gram-negative bacterial infections. Current FDA-approved prescribing information recommends that polymyxin B dosing should be adjusted according to patient's renal function, despite studies have shown poor correlation between creatinine and polymyxin B clearance. The objective of the study was to determine if steady state polymyxin B exposures in patients with normal renal function were different from those with renal insufficiency. Nineteen adult patients who received intravenous polymyxin B (1.5 - 2.5 mg/kg actual body weight daily) were included. To measure polymyxin B concentrations, serial blood samples were obtained from each patient after receiving polymyxin B for at least 48 hours. The primary outcome was polymyxin B exposure at steady state, as reflected by the area under the concentration-time curve (AUC) over 24 hours. Five patients had normal renal function [estimated creatinine clearance (CrCL) ≥ 80 ml/min] at baseline whereas 14 had renal insufficiency (CrCL< 80 ml/min). Mean ± SD AUC of polymyxin B in the normal renal function cohort was 63.5 ± 16.6 mg.h/l, as compared to 56.0 ± 17.5 mg.h/l in renal insufficiency cohort ( p = 0.42). Adjusting the AUC for the daily dose (in mg/kg of actual body weight) did not result in a significant difference (28.6 ± 7.0 mg.h/l vs, 29.7 ± 11.2 mg.h/l, p = 0.80). Polymyxin B exposures in patients with normal and impaired renal function after receiving standard dosing of polymyxin B were comparable. Polymyxin B dosing adjustment in patients with renal insufficiency should be re-examined.
Article
Full-text available
Polymyxin B is increasingly used as a treatment of last resort for multidrug-resistant Gram-negative infections. Despite being available as a mixture of several structurally related analogues, the properties are commonly reported as an aggregate of the individual components. We compared the pharmacokinetics of individual polymyxin B components in an animal model and in humans. There were no considerable differences observed in the pharmacokinetics among major components of polymyxin B. Combining different components for pharmacokinetic analysis appeared reasonable.
Article
Full-text available
Despite dose-limiting nephrotoxicity concerns, polymyxin B has resurged as the last treatment resort for multidrug-resistant Gram-negative bacterial infections. However, the pharmacokinetic, pharmacodynamic and nephrotoxic properties of polymyxin B are still not thoroughly understood. The objective of this study was to provide additional insights to the overall bio-distribution and disposition of polymyxin B in an animal model. Sprague-Dawley rats were dosed with intravenous polymyxin B (3 mg/kg). Drug concentrations in the serum, urine, bile and tissue (brain, heart, lungs, liver, spleen, kidneys and skeletal muscle) samples over time were assayed by a validated methodology. Among all the organs evaluated, polymyxin B distribution was the highest in the kidneys. The mean renal tissue/serum polymyxin B concentration ratios were 7.45 (95% CI, 4.63-10.27) at 3 h and 19.62 (95% CI, 5.02-34.22) at 6 h post-dose, respectively. Intrarenal drug distribution was examined by immunostaining. Using a ratiometric analysis, proximal tubular cells showed the highest accumulation of polymyxin B (Mander's overlap coefficient 0.998), among all other cell types evaluated. Less than 5% of the administered dose was recovered in urine over 48 h, but all 4 major polymyxin B components were detected in the bile over 4 h. These findings corroborate previous results that polymyxin B is highly accumulated in the kidneys, but the elimination is likely via a non-renal route. Biliary excretion could be one of the possible routes of polymyxin B elimination, which should be further explored. Elucidation of mechanism(s) of drug uptake in proximal tubular cells is ongoing.
Article
Full-text available
Introduction: In the era of multidrug-resistant, extensively drug-resistant (XDR) and even pandrug-resistant Gram-negative microorganisms, the medical community is facing the threat of untreatable infections particularly those caused by carbapenemase-producing bacteria, that is, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii. Therefore, all the presently available antibiotics, as well as for the near future compounds, are presented and discussed. Areas covered: Current knowledge concerning mechanisms of action, in vitro activity and interactions, pharmacokinetic/pharmacodynamics, clinical efficacy and toxicity issues for revived and novel antimicrobial agents overcoming current resistance mechanisms, including colistin, tigecycline, fosfomycin, temocillin, carbapenems, and antibiotics still under development for the near future such as plazomicin, eravacycline and carbapenemase inhibitors is discussed. Expert opinion: Colistin is active in vitro and effective in vivo against XDR carbapenemase-producing microorganisms in the critically ill host, whereas tigecycline, with the exception of P. aeruginosa, has a similar spectrum of activity. The efficacy of combination therapy in bacteremias and ventilator-associated pneumonia caused by K. pneumoniae carbapenemase producers seems to be obligatory, whereas in cases of P. aeruginosa and A. baumannii its efficacy is questionable. Fosfomycin, which is active against P. aeruginosa and K. pneumoniae, although promising, shares poor experience in XDR infections. The in vivo validity of the newer potent compounds still necessitates the evaluation of Phase III clinical trials particularly in XDR infections.
Article
Full-text available
Colistin and polymyxin B have indistinguishable microbiological activity in vitro, but they differ in the form administered parenterally to patients. Polymyxin B is administered directly as the active antibiotic, whereas colistin is administered as the inactive prodrug, colistin methanesulfonate (CMS). CMS must be converted to colistin in vivo, but this occurs slowly and incompletely. Here we summarize the key differences between parenteral CMS/colistin and polymyxin B, and highlight the clinical implications. We put forth the view that overall polymyxin B has superior clinical pharmacological properties compared with CMS/colistin. We propose that in countries such as the United States where parenteral products of both colistin and polymyxin B are available, prospective studies should be conducted to formally examine their relative efficacy and safety in various types of infections and patients. In the meantime, where clinicians have access to both polymyxins, they should carefully consider the relative merits of each in a given circumstance.
Article
Full-text available
Despite concerns of nephrotoxicity, polymyxin antibiotics often remain the only susceptible agents for multidrug-resistant (MDR) Gram-negative bacteria. Colistin has been more commonly used clinically due to a perceived safety benefit. We compared the nephrotoxicity of colistin to polymyxin B. The in vitro cytotoxicity of colistin was compared to polymyxin B in two mammalian renal cell lines. To validate the clinical relevance of the findings, we evaluated adult patients with normal renal function who received a minimum of 72 h of polymyxin therapy in a multicenter study. The primary outcome was the prevalence of nephrotoxicity, as defined by the RIFLE (risk, injury, failure, loss, end-stage kidney disease) criteria. Colistin exhibited an in vitro cytotoxicity profile similar to polymyxin B. A total of 225 patients (121 receiving colistimethate, 104 receiving polymyxin B) were evaluated. Independent risk factors for colistimethate-associated nephrotoxicity included age (odds ratio [OR], 1.04; 95% confidence interval [CI], 1.00 to 1.07; P = 0.03), duration of therapy (OR 1.08; 95% CI, 1.02 to 1.15; P = 0.02), and daily dose by ideal body weight (OR 1.40; 95% CI, 1.05 to 1.88; P = 0.02). In contrast, cystic fibrosis was found to be a protective factor in patients who received colistimethate (OR, 0.03; 95% CI, 0.001 to 0.79; P = 0.04). In a matched analysis based on the risk factors identified (n = 76), the prevalence of nephrotoxicity was higher with colistimethate than with polymyxin B (55.3% versus 21.1%; P = 0.004). Polymyxin B was not found to be more nephrotoxic than colistin and may be the preferred polymyxin for MDR infections. A prospective study comparing the two polymyxins directly is warranted.
Article
Full-text available
Enterobacteriaceae showing resistance to cephalosporins due to extended-spectrum β-lactamases (ESBLs) or plasmid-mediated AmpC enzymes, and those producing carbapenemases have spread worldwide during the last decades. Many of these isolates are also resistant to other first-line agents such as fluoroquinolones or aminoglycosides, leaving few available options for therapy. Thus, older drugs such as colistin and fosfomycin are being increasingly used. Infections caused by these bacteria are associated with increased morbidity and mortality compared with those caused by their susceptible counterparts. Most of the evidence supporting the present recommendations is from in vitro data, animal studies, and observational studies. While carbapenems are considered the drugs of choice for ESBL and AmpC producers, recent data suggest that certain alternatives may be suitable for some types of infections. Combined therapy seems superior to monotherapy in the treatment of invasive infections caused by carbapenemase-producing Enterobacteriaceae. Optimization of dosage according to pharmacokinetics/pharmacodynamics data is important for the treatment of infections caused by isolates with borderline minimum inhibitory concentration due to low-level resistance mechanisms. The increasing frequency and the rapid spread of multidrug resistance among the Enterobacteriaceae is a true and complex public health problem.
Article
Full-text available
Background: Polymyxin B is a last-line therapy for multidrug-resistant gram-negative bacteria. There is a dearth of pharmacokinetic data to guide dosing in critically ill patients. Methods: Twenty-four critically ill patients were enrolled and blood/urine samples were collected over a dosing interval at steady state. Polymyxin B concentrations were measured by liquid chromatography-tandem mass spectrometry. Population pharmacokinetic analysis and Monte Carlo simulations were conducted. Results: Twenty-four patients aged 21-87 years received intravenous polymyxin B (0.45-3.38 mg/kg/day). Two patients were on continuous hemodialysis, and creatinine clearance in the other patients was 10-143 mL/min. Even with very diverse demographics, the total body clearance of polymyxin B when scaled by total body weight (population mean, 0.0276 L/hour/kg) showed remarkably low interindividual variability (32.4% coefficient of variation). Polymyxin B was predominantly nonrenally cleared with median urinary recovery of 4.04%. Polymyxin B total body clearance did not show any relationship with creatinine clearance (r(2) = 0.008), APACHE II score, or age. Median unbound fraction in plasma was 0.42. Monte Carlo simulations revealed the importance of initiating therapeutic regimens with a loading dose. Conclusions: Our study showed that doses of intravenous polymyxin B are best scaled by total body weight. Importantly, dosage selection of this drug should not be based on renal function.
Article
Polymyxin B (PB) is an antibiotic consisting of a cyclic heptapeptide and a tripeptide side chain used in treatment of infections caused by Gram-negative bacteria. Commercial formulations of PB contain multiple structurally related components with major constituents of PB1, PB2, PB3 and ile-PB1. To understand the pharmacokinetics of these major components, we have developed and validated a LC-MS/MS method to quantify PB1, PB2, PB3 and ile-PB1 in human plasma. PB was extracted from plasma by protein precipitation using trichloroacetic acid followed by chromatographic separation on Zorbax Bonus-RP column (100 mm × 2.1 mm, 1.8 μm) using stepwise gradient elution of water containing 0.1% of formic acid and 0.1% of trichloroacetic acid (mobile phase A) and 90% acetonitrile with 0.1% formic acid (mobile phase B). Despite of structural similarities, these PBs were completely resolved in the analytical run time of 6.5 min. Detection and quantification of PBs were performed by selected reaction monitoring (SRM) under positive ionization mode in the mass spectrometer. Separation of PB1 and ile-PB1, as well as PB2 and PB3, before quantification is crucial because they are structural isomers detected based the same SRM. Excellent linearity was achieved (r² > 0.99) in the calibration curves of PB. The developed method was accurate (95.3–111.7%) and precise (CV < 5.1%). Recovery of PB from the plasma extraction was between 53–76% and reproducible (CV < 4.5%). Matrix effect was not observed by post-column infusion of PB in the mass spectrometer. This methodology has been successfully applied to clinical study of patients dosed with intravenous infusions of PB.