You are on page 1of 9

Curr Infect Dis Rep (2017) 19:38

DOI 10.1007/s11908-017-0596-3

ANTIMICROBIAL DEVELOPMENT AND DRUG RESISTANCE (A PAKYZ, SECTION EDITOR)

Polymyxin Resistance in Gram-negative Pathogens


Pavithra Srinivas 1 & Kaitlyn Rivard 1

# Springer Science+Business Media, LLC 2017

Abstract Introduction
Purpose of Review The polymyxins are one of the last line
antimicrobial classes to retain activity against multidrug- The emergence of multidrug-resistant (MDR) Gram-negative
resistant Gram-negative bacilli. However, with the increased organisms combined with the lack of novel antimicrobials has
use of polymyxins in recent years, reports of resistance have been recognized worldwide as a significant concern [1]. MDR
also been increasing. We aimed to describe the mechanisms organisms such as Acinetobacter baumannii and
and occurrence of polymyxin resistance in Gram-negative or- Pseudomonas aeruginosa harbor multiple resistance mecha-
ganisms and propose strategies to overcome resistance. nisms, thereby limiting treatment options. The polymyxins,
Recent Findings The most common mechanism of acquired colistin (polymyxin E), and polymyxin B remain one of the
resistance to the polymyxins is via modification of the bacte- last classes of antimicrobials to retain activity against these
rial outer membrane lipopolysaccharide. Global epidemiolog- MDR Gram-negative organisms [2]. The polymyxins are
ical surveillance studies have reported the occurrence of poly- lipopeptide antibiotics, isolated from the Gram-positive or-
myxin resistance to be most common in Enterobacteriaceae, ganism Paenibacillus polymyxa (previously Bacillus
specifically Enterobacter species and Acinetobacter polymyxa) in the 1940s, that exert bactericidal activity against
baumannii. Prevalence of polymyxin-resistant Gram-negative Gram-negative bacilli by disrupting the bacterial cell mem-
organisms varies significantly by geographical location. brane via both electrostatic and hydrophobic interactions. By
Summary Emergence of polymyxin resistance is of great con- binding to the lipopolysaccharides (LPS) in the outer mem-
cern, given the limited number of agents available to treat brane of Gram-negative bacilli, the positively charged poly-
infections caused by multidrug-resistant Gram-negative or- myxin molecule interacts with the negatively charged phos-
ganisms. Strategies to mitigate the development of polymyxin phate groups on the lipid A moiety of the LPS, thereby
resistance include dose optimization and using polymyxin destabilizing the outer membrane. As a result of this destabi-
agents in combination with other highly active antimicrobial lization, the polymyxin molecule is able to insert its hydro-
agents. phobic regions within the outer membrane causing further
membrane disruption and facilitating further uptake of poly-
Keywords Colistin . Colistimethate sodium . Polymyxin B . myxins [3].
Multidrug-resistant gram-negative bacilli The polymyxins initially fell out of favor due to concerns
surrounding nephrotoxic and neurotoxic adverse effects.
This article is part of the Topical Collection on Antimicrobial However, with the emergence of MDR Gram-negative organ-
Development and Drug Resistance isms, there has been an increased utilization of these agents in
recent years [2]. Along with increased use of polymyxins,
* Pavithra Srinivas there have also been increasing reports of resistance to the
srinivp2@ccf.org polymyxins [4, 5, 6•, 7–9]. The purpose of this review is to
describe the mechanisms and occurrence of polymyxin resis-
1
Department of Pharmacy, Cleveland Clinic, 9500 Euclid Ave tance in Gram-negative organisms and propose strategies to
(Hb-105), Cleveland, OH 44195, USA overcome resistance.
38 Page 2 of 9 Curr Infect Dis Rep (2017) 19:38

Mechanisms of Polymyxin Resistance Synthesis of PEtN is also mediated through the PhoP/PhoQ
and PmrA/PmrB TC pathway. Once PmrA/PmrB is
The most common mechanism for the development of resis- expressed, pmrC (also known as eptA) and cptA (also known
tance to the polymyxins is via modification of the bacterial as eptC) are activated to synthesize PEtN for addition to the
outer membrane LPS. In this section, we describe the various lipid A or Kdo components of the LPS [10, 11]. EptB (also
mechanisms involved in the development of polymyxin known as pagC) also results in modification of the LPS with
resistance. PEtN. MgrR acts as a negative feedback regulator for eptB.
Activation of mgrR by the PhoP/PhoQ TCs leads to repression
Intrinsic Resistance of eptB and decreased PEtN production [14]. Similar to the L-
Ara4N pathway, mutations in any of these genes or TCs may
The polymyxins have no activity against anaerobes and lead to dysregulation and overproduction of PEtN, which
Gram-positive organisms, as they lack the presence of an changes the charge of the LPS.
LPS containing outer cell membrane. A number of Gram- Additional methods for LPS modification have been dis-
negative organisms are naturally resistant to polymyxins, in- covered including genes that result in acylation and
cluding Brucella spp., Burkholderia cepacia complex, deacylation of lipid A (IpxM, pagL, IpxR), additional TCs
Edwardsiella spp., Morganella morganii, Proteus spp., resulting in L-Ara4N synthesis and addition to the LPS
Providencia spp., and Serratia spp. The mechanism of this (colR/colS, cprR/cprS, parR/parS), and complete loss of the
natural resistance is hypothesized to be due to constitutive LPS (IpxA, IpxC, IpxD) [10]. It is important to note that the
gene expression resulting in the addition of cationic molecules mechanism of LPS modification differs between Gram-
to the LPS, leading to decreased binding affinity at the poly- negative bacterial species.
myxin site of action on the LPS [10–12].
Plasmid-mediated mcr-1 Gene
Modification of the LPS
Modifications of the lipid A or Kdo components of the LPS in
Similar to naturally resistant Gram-negative pathogens, organ- Gram-negative organisms via the PmrA/PmrB, PhoP/PhoQ,
isms that are susceptible to polymyxins in vitro may acquire and mgrB/mgrR inactivation pathways are chromosomally
mutations in cellular pathways that lead to modification of the mediated, precluding the horizontal transfer of resistance
LPS. Modification primarily occurs via the addition of cation- genes between bacterial generator species. However, recent
ic molecules, 4-amino-4-deoxy-L-arabinose (L-Ara4N), and reports have identified a new colistin resistance gene, mcr-1,
phosphoethanolamine (PEtN) to the lipid A or 3-deoxy-D- encoding a PEtN transferase [10]. The mcr-1 gene was first
mannooctulosonic acid (Kdo) components of the LPS. identified in commensal Escherichia coli isolates from food
The arnBCADTEF (also known as the pmrHFIJKLM) op- animals in China during surveillance [15••]. However, within
eron and the pmrE operon, when activated, synthesize L- months, dissemination of the gene in food animals in multiple
Ara4N from uridine diphosphate glucuronic acid to be added countries around the globe was reported [16]. Although most
to the LPS [10, 11]. The arnBCADTEF and pmrE operons are mcr-1-mediated colistin resistance has been isolated in
regulated by a pathway of two bacterial two-component sys- carbapenem-susceptible organisms, the plasmid-mediated na-
tems (TCs), PhoP/PhoQ and PmrA/PmrB. Activation of the ture of the gene raises concerns for inter-species transfer of
PhoP/PhoQ TCs leads to activation of pmrD, which in turn colistin resistance among Gram-negative organisms [10, 17].
activates the PmrA/PmrB TCs. PmrA/PmrB then up-regulates The mcr-1 gene has also been reported in species of
the arnBCADTEF and pmrE operons to synthesize L-Ara4N Salmonella, Shigella, Klebsiella, and Enterobacter [10].
for addition to the LPS [10, 11]. In some bacterial Gram- Amino acid sequencing of the mcr-1 gene has revealed a
negative species, such as Klebsiella spp., the PhoP/PhoQ close association with PEtN transferases (pmrC) found in
TCs can also directly activate the arnBCADTEF and pmrE Paenibacillus spp., the bacteria that produces polymyxins.
operons independent of pmrD and PmrA/PmrB [10]. MgrB Structural analysis of protein sequencing revealed the pres-
acts as negative feedback regulator for PhoP/PhoQ and sup- ence of two PEtN transferases, LptA from Neisseria
presses the L-Ara4N synthesis pathway [13]. Mutations in any meningitidis and eptC (or cptA) from Campylobacter jejuni,
of the aforementioned genes or TCs may lead to dysregulation both of which are intrinsically resistant to polymyxins [15••].
and overproduction of L-Ara4N synthesis. Additionally, envi-
ronmental stimuli can promote the activation of these systems Heteroresistance
[3, 11]. Incorporation of the overproduced L-Ara4N into the
LPS leads to a net positive charge on the bacterial cell mem- Heteroresistance is defined as the presence of subpopulations
brane, impairing the ability of the polymyxins to bind to their of resistant organisms in an isolate considered to be suscepti-
site of action. ble by standard testing methodologies. It is a poorly
Curr Infect Dis Rep (2017) 19:38 Page 3 of 9 38

characterized phenomenon wherein a population of bacteria MIC ≥ 4 mg/L meets the epidemiologic cutoff for resistance
with similar genotypes may exhibit variable phenotypic re- [27]. As such, reported rates of colistin and polymyxin B
sponses to an antimicrobial agent. Heteroresistance may also resistance range widely across the globe.
be described as heterogeneous resistance, population-wide
variation of resistance, and heterogeneity of response to anti- Acinetobacter baumannii
biotics. It was first described in 1947 for Haemophilus
influenzae, followed by Staphylococcus spp. in the 1970s, Global rates of polymyxin-resistant Acinetobacter spp.
and more recently for Gram-negative bacilli with polymyxins. have been steadily increasing since the first description
The clinical significance of heteroresistance is still unclear, but of a colistin-resistant strain in 1999. Reported rates of
the concern remains that the more resistant subpopulations colistin- and polymyxin B-resistant A. baumannii in the
may be selected out during therapy [18]. worldwide SENTRY Antimicrobial Surveillance database
Further complicating the phenomenon is the ability to mea- from 2001 to 2011 ranged from 0.9 to 3.3% [4, 5].
sure heteroresistance in vitro. The gold standard for determin- When stratified by geographic location, rates of poly-
ing heteroresistance is a population analysis profile (PAP) myxin resistance have been higher in the US (0.8 to
methodology, wherein a bacterial population is exposed to a 4.8%) compared to Europe (0.4 to 2.7%), Latin
gradient of antibiotic concentrations and bacterial growth at America (0.9 to 1.1%), and the Asia-Pacific (0.7 to
each concentration is quantified. The pitfalls of PAPs are that 1.7%) regions [5, 6•]. Additionally, surveillance data
they are labor intensive, time-consuming processes that may from 2006 to 2009 demonstrated higher rates of resis-
not be suitable for clinical laboratories to perform [18]. tance to polymyxin B among imipenem-non-susceptible
Heteroresistance to colistin was first described in 2006 strains of A. baumannii (2.1%) in US hospitals, com-
among clinical isolates of multidrug-resistant A. baumannii pared to imipenem-susceptible strains (0.8%) [5]. Other
that were reported as susceptible to colistin based on the min- case reports from Europe have observed general rates of
imum inhibitory concentration (MIC) [19]. The authors raised colistin resistance in A. baumannii of < 7%, except two
concerns for rapid development of resistance and therapeutic reports from Bulgaria and Spain demonstrating rates of
failure if colistin is used at suboptimal doses or as monother- 16.7 and 19.1%, respectively [4, 29, 30]. Data from a
apy. A subsequent study attempted to associate colistin European surveillance network (EARS-Net) comprising
heteroresistance in A. baumannii with prior colistin therapy of 20 European countries from 2014 showed an overall
and demonstrated a significantly higher degree of 4% rate of polymyxin resistance in Acinetobacter spp.,
heteroresistance in isolates from patients with prior colistin with the majority (80.1%) of them reported from Greece
treatment [20]. Colistin and polymyxin B heteroresistance and Italy [31].
have also been described in various other Gram-negative or- On the other hand, heteroresistance rates to colistin in
ganisms (Table 1). However, the majority of the literature is A. baumannii have ranged from 14 to 100% in reports from
limited by small sample sizes, varying methodologies for de- various parts of the world. Most of these reports were com-
termining heteroresistance, and large variability in the rates of prised of small sample sizes and varying testing methodolo-
heteroresistance detected [19–26]. gies, likely explaining the higher incidence and larger varia-
tion of reported rates [19–26]. The primary mechanism of
polymyxin resistance in A. baumannii is via mutations in the
Epidemiology of Polymyxin Resistance PmrA/PmrB genes leading to the addition of PEtN to the
bacterial outer membrane LPS, as A. baumannii lacks the
The Clinical and Laboratory Standards Institute (CLSI) and arnBCADTEF and pmrE operons required to synthesize L-
European Committee on Antimicrobial Susceptibility Testing Ara4N [3, 32].
(EUCAST) have set the susceptibility breakpoint for colistin
and polymyxin B as ≤ 2 mg/L for A. baumannii and Pseudomonas aeruginosa
P. aeruginosa and the epidemiological breakpoint as ≤ 2 mg/
L for Enterobacteriaceae [27, 28]. However, definitions of Polymyxin resistance in P. aeruginosa is predominantly
resistance are slightly different between the CLSI and developed by the addition of L-Ara4N to bacterial outer
EUCAST. The EUCAST guidelines consider any organism membrane LPS, via PhoP/PhoQ-mediated activation of
with a colistin MIC > 2 mg/L to be resistant [28]. arnBCADTEF [3, 32]. While the polymyxins have main-
Alternatively, the CLSI guidelines provide organism-specific tained excellent activity against P. aeruginosa over the
resistance cutoffs. For A. baumannii, a colistin/polymyxin B years, the increased use of inhaled colistin, particularly
MIC ≥ 4 mg/L is considered resistant; for P. aeruginosa, a in cystic fibrosis patients, has led to the emergence of
colistin MIC ≥ 4 mg/L or a polymyxin B MIC ≥ 8 mg/L is resistance [7]. SENTRY Antimicrobial Surveillance data
considered resistant; and for Enterobacteriaceae, a colistin from US and European hospitals evaluated between 2009
38 Page 4 of 9 Curr Infect Dis Rep (2017) 19:38

Table 1 Prevalence of heteroresistance to colistin or polymyxin B among Gram-negative bacilli

Organism Antibiotic Detection of Year Comments Reference


heteroresistance

Acinetobacter baumannii Colistin 15/16 (93.7%) 2006 Some subpopulations were able to grow in the presence of up to [19]
200 μg/mL of colistin
Acinetobacter Colistin 19/19 (100%) 2008 Significantly higher degree of heteroresistance found among isolates [20]
baumannii-- from patients with previous colistin treatment. All isolates maintained
calcoaceticus complex colistin MICs > 16 mg/L after two passages on sheep blood plates
Acinetobacter baumannii Colistin 13/28 (46.4%) 2009 Case report of a patient with post-neurosurgical meningitis with [22]
development of colistin resistance during intrathecal treatment
Acinetobacter baumannii Colistin 1/7 (14%) 2007 All isolates maintained resistance to colistin after passaging on sheep [21]
Enterobacter cloacae 6/10 (60%) blood agar and retesting, suggesting resistance is induced upon
exposure to colistin rather than via a stable mutation
Enterobacter cloacae Polymyxin 8/8 (100%) 2013 In several isolates, the number of heteroresistant colonies was greater at [25]
B higher concentrations of polymyxin B (4–8 μg/mL) compared to
lower concentrations (0.5–2 μg/mL)
Carbapenemase-producing Colistin 12/16 (75%) 2011 All heteroresistant strains except one maintained colistin MICs [26]
Klebsiella pneumoniae > 8 mg/L after serial passages on colistin-free media
Pseudomonas aeruginosa Polymyxin 1/24 (4%) 2013 Rate of heterogeneous subpopulations with increased MICs to [24]
B polymyxin B but still < 2 mg/L were 37.5% (9/24)
Burkholderia cenocepacia Polymyxin – 2013 More resistant subpopulations of B. cenocepacia may communicate [23]
B high level resistance to less resistant cells

to 2012 have reported rates of colistin resistance in Klebsiella spp. and 0.7% in ESBL E. coli from European
P. aeruginosa around 0.3% [6•]. Worldwide reports of hospitals [6•]. In a recent global surveillance program of
polymyxin resistance rates in P. aeruginosa have similar- 19,719 isolates of Enterobacteriaceae collected between
ly ranged from 0.1 to 0.6% [5]. However, surveillance 2012 and 2013 from 39 countries, the highest rates of
data of P. aeruginosa isolates from Canadian hospitals polymyxin resistance occurred in Enterobacter asburiae
from 2008 to 2015 demonstrated overall colistin resis- (39.1%), Enterobacter cloacae (3%), and Klebsiella
tance rates of 5.1%, with resistance rates in MDR and pneumoniae (2.4%) isolates. Regional distribution of
extensively drug resistant (XDR) isolates ranging from polymyxin resistance was similar worldwide—Europe
7.1 to 11.7%. One proposed reason for the higher rates (1.8%), Latin America (1.5%), Middle East-Africa
of colistin resistance reported is that patients with cystic (1.4%), North America (1.3%), and Asia-Pacific (1.3%).
fibrosis and other conditions predisposing to multidrug Alarmingly, polymyxin resistance was significantly
resistance were not excluded from the Canadian surveil- higher in carbapenemase-producing Enterobacteriaceae,
lance database [8]. with rates nearing 12%, suggesting a strong association
between the presence of a carbapenemase and decreased
Enterobacteriaceae activity of polymyxin against these isolates.
K. pneumoniae was the most common polymyxin-resis-
Among the Enterobacteriaceae, Klebsiella spp. is the tant, carbapenemase-producing species [9].
most common genus associated with polymyxin resis- While most Enterobacteriaceae develop resistance to the
tance. However, recent surveillance data from 2009 to polymyxins via chromosomal modifications to the outer mem-
2012 has also demonstrated high polymyxin resistance brane LPS, the predominant pathway utilized is organism spe-
rates in Enterobacter spp., ranging from 13.6 to 15%, in cific. For example, Klebsiella spp. rely more on the addition of
US and European hospitals. Rates of polymyxin resis- L-Ara4N to the lipid A via mutations in the mgrB negative
tance in Klebsiella spp. have been reported at 2.7 and feedback regulator of PhoP/PhoQ, whereas E. coli develops
3.6%, in the US and European regions, respectively, resistance via the addition of PEtN to the LPS core due to
while resistance rates in E. coli have been significantly mutations in the mgrR negative feedback regulator and eptB
lower at 0.3 and 0.1%, respectively. Extended-spectrum [10, 32]. More recently, the plasmid mediated mcr-1 gene has
beta-lactamase producing (ESBL) Klebsiella spp. and also been implicated in colistin-resistance among E. coli [17].
E. coli have demonstrated higher rates of resistance to The first report of a pathogenic E. coli harboring the mcr-1 gene
polymyxins—11.5% in ESBL Klebsiella spp. and 1.4% was identified in the urine culture of a patient in Pennsylvania
in ESBL E. coli from US hospitals and 7.8% in ESBL in April 2016 [33]. Since then, the mcr-1 gene has been
Curr Infect Dis Rep (2017) 19:38 Page 5 of 9 38

identified in human isolates from eight additional states in the achievement of clinically significant plasma concentrations.
USA as well as in case reports from Italy and Egypt [34–36]. However, this rapid renal elimination results in high urinary
concentrations of colistin, as CMS continues to be converted
to active drug in the urine. Furthermore, significant inter-
Therapeutic Approaches to Combating Polymyxin patient variability in plasma colistin Cssavg in the tenfold range
Resistance raises additional concerns for achieving drug concentrations
that are both effective and safe, due to the narrow therapeutic
The two polymyxin agents used in clinical practice, colistin window of colistin and increased risk of nephrotoxicity at
and polymyxin B, share similarities in mechanism of action concentrations > 2.5 mg/L [40–42••].
and spectrum of activity. They differ structurally only in one Polymyxin B, on the other hand, can rapidly achieve
amino acid and in commercial availability. Colistin is com- desired plasma concentrations with less inter-patient var-
mercially available as the inactive prodrug, colistimethate so- iability, in the range of only three- to fourfold at various
dium (CMS), which requires activation in vivo via hydrolysis. renal functions, due to being administered as the active
Polymyxin B, on the other hand, is available as the active drug [37, 42••, 43]. However, polymyxin B is eliminated
sulfate salt form, which begets significant pharmacokinetic via non-renal mechanisms and does not achieve thera-
differences in vivo [37]. Both agents are available as parenter- peutic concentrations in the urinary tract.
al formulations; however, availability of each agent varies by Despite pharmacokinetic differences between CMS
country. For example, some countries such as Japan and South and polymyxin B, clinical comparisons of efficacy and
Africa do not have access to either of the polymyxins, whereas toxicities are limited. Most studies comparing colistin
in Europe and Australia, the CMS formulation is the only and polymyxin B have focused on nephrotoxicity, with
commercially available product. In the USA and a number efficacy outcomes being secondary. Furthermore, these
of other countries, both formulations are available [3, 37]. studies have been retrospective and limited in sample
The pharmacokinetic/pharmacodynamic (PK/PD) parame- size. Oliveira and colleagues were the first to compare
ter best associated with polymyxin activity is the fractional clinical outcomes between colistin and polymyxin B in
area under the curve of the drug over the MIC of the organism 82 patients and concluded that both agents were compa-
(fAUC/MIC). It is proposed that the average steady-state con- rable with regard to nephrotoxicity and efficacy [44].
centration (Css,avg) of drug should be greater than or equiva- Other comparative studies have also observed no signif-
lent to the pathogen’s MIC to ensure optimal antibacterial icant differences in in-hospital and 30-day mortality
activity [38, 39]. Therefore, based on CLSI and EUCAST rates between colistin and polymyxin B, although differ-
susceptibility breakpoints, it would prudent to target a colistin ences in nephrotoxicity outcomes have varied [45–48].
or polymyxin B plasma Css,avg of at least 2 mg/L empirically, While clinical efficacy may not be significantly differ-
until confirmation of pathogen MICs. ent, recent reports of nephrotoxicity may favor the use
Appropriate utilization of the polymyxins is key to over- of polymyxin B over colistin. Although literature com-
coming or preventing polymyxin resistance. A thorough un- paring nephrotoxicity rates between colistin and poly-
derstanding of the PK/PD properties of the polymyxins is myxin B are limited to six studies, rates of acute kidney
needed to determine the appropriate agent to use in a given injury (AKI) reported have ranged widely from 25 to
situation (when both formulations are available), select an 60% with CMS and 21–41% with polymyxin B
appropriate dose, and use effectively in combination with oth- [44–49]. Studies performed more recently have reported
er antimicrobial agents. higher rates of AKI compared to older studies, likely a
reflection of updated, more aggressive dose recommen-
Polymyxin Agent Selection dations for colistin and polymyxin B.
As such, we recommend that polymyxin B be considered
Colistin’s commercial availability as the prodrug form, CMS, over CMS for the treatment of serious systemic infections
presents challenges in its ability to rapidly and reliably reach given its more reliable pharmacokinetic profile. However,
target steady-state plasma concentrations. Once administered, due to the inability of polymyxin B to achieve adequate con-
CMS undergoes slow and variable hydrolysis into colistin, centrations in the urine, CMS may still be considered for uri-
leading to delayed achievement of therapeutic concentrations nary tract infections [37, 43]. Additionally, colistin remains
of active drug [37, 40]. CMS has a relatively short half-life (2– the preferred agent for inhalation therapy due to reports
3 h) and undergoes rapid renal elimination, whereas the active of bronchoconstriction with inhaled polymyxin B [50]. If
colistin has a longer half-life (14 h) and is eliminated via non- polymyxin B is unavailable, CMS may still be considered
renal pathways. Due to its rapid renal clearance, it is estimated for systemic infections. Regardless of which agent is uti-
that only 20–25% of CMS is converted to colistin systemical- lized, dose-optimization is essential to minimize the de-
ly in patients with normal renal function, thereby delaying velopment of resistance.
38 Page 6 of 9 Curr Infect Dis Rep (2017) 19:38

Dosing Optimization recommendations are supported by a limited amount of liter-


ature. Similar to CMS, dosing for polymyxin B may be
Since the polymyxins were developed before the Food and expressed in terms of IUs or mg—10,000 IUs is equivalent
Drug Administration (FDA) developed standards for approval to 1 mg of polymyxin B. The FDA-recommended polymyxin
of new drugs, they did not undergo rigorous evaluation of B dose is 1.5–2.5 mg/kg/day, calculated from total body
dosing for efficacy and safety. As such, variations exist in weight and divided every 12 h [43]. Although the product
colistin dosing recommendations between the FDA, the package insert recommends a dose reduction in patients with
European Medicines Agency (EMA), and population renal impairment, recent pharmacokinetic data has suggested
pharmacokinetics-based data [42••, 51, 52]. The FDA recom- that polymyxin B doses are better scaled by total body weight
mends weight-based dosing determined by ideal body weight rather than renal function [54]. Sandri et al. also attempted to
and adjusted for renal function. The EMA recommends a flat provide guidance regarding optimal polymyxin B dosing in
CMS dose based on a calculated Cockroft-Gault creatinine their pharmacokinetic analysis of 24 patients. Based on Monte
clearance, along with a loading dose for critically ill patients. Carlo simulations of polymyxin B exposures for various dos-
Nation and colleagues also recommend initiating colistin ther- ing regimens, the regimen with the highest probability of tar-
apy with a loading dose; however, they recommend a CMS get fAUC/MIC and Css,avg concentrations was a 2.5-mg/kg
dosing equation that incorporates the target Css,avg and renal loading dose followed by 1.5 mg/kg every 12 h. However,
function to derive a dose [42••, 53••]. To complicate matters as seen with colistin, for pathogens with MICs ≥ 2 mg/L, the
further, CMS dosing is expressed in different units depending probability of target fAUC/MIC and Css,avg attainment was
on the region of the world, international units (IU), or colistin significantly lower [54]. Therefore, combination antimicrobial
base activity (CBA). One million IUs of CMS is equivalent to therapy is warranted in patient populations with decreased
approximately 30-mg CBA. likelihood of achieving goal colistin/polymyxin B concentra-
Comparisons of the three colistin dosing recommendations tions, such as patients with good renal function or infections
(FDA, EMA, and Nation et al.) and their respective probabil- with organisms with MICs > 0.5 mg/L.
ities of achieving target PK/PD parameters for various MICs At this time, there is no “one size fits all” dosing recom-
have demonstrated that all three dosing schemes would per- mendation for the polymyxins, as more data are needed to
form well when treating a pathogen with a colistin MIC of truly understand the best dose to maximize efficacy and min-
0.5 mg/L (Table 2) [40, 42••, 53••]. However, with rising imize toxicity. We recommend consideration of the pathogen
colistin MIC, the ability to achieve a Css,avg ≥ 1 mg/L in MIC, patient-specific factors, and evaluation of the most up-
patients with normal renal function (CrCl > 80 ml/min) drops dated literature to determine the optimal dosing regimen for
significantly, likely due to the rapid clearance of CMS pre- either agent for a given patient.
cluding conversion to active colistin in vivo. In patients with
impaired renal function (CrCl < 80 ml/min), FDA dosing rec- Combination Therapy
ommendations often fail to achieve target Css,avg ≥ 2 mg/L,
whereas dosing regimens proposed by the EMA and Nation Various pharmacokinetic and clinical evaluations have asso-
et al. are able to reasonably achieve appropriate Css,avg. ciated colistin or polymyxin B monotherapy with suboptimal
Polymyxin B, on the other hand, is administered in its outcomes. An in vitro pharmacodynamics study of polymyxin
active form and drug clearance is not impacted by renal func- B against four strains of P. aeruginosa demonstrated a signif-
tion. While dosing of polymyxin B is less ambiguous than icant reduction in bactericidal activity with a high inoculum
CMS, it is important to note that polymyxin B dosing and selective amplification of resistant subpopulations at 24 h

Table 2 Percent of patients achieving goal Css,avg by dosing strategy [42••, 53••]

CrCl (mL/min) FDA dosing EMA dosing Nation et al. dosing

Css,avg Css,avg Css,avg Css,avg Css,avg Css,avg Css,avg Css,avg Css,avg Css,avg Css,avg Css,avg
0.5 1 2 4 0.5 1 2 4 0.5 1 2 4

≥ 80 > 90% 60–70% 20–30% 0–10% 100% 70–80% 30–40% 0–10% 100% 70–80% 30–40% 0–10%
50–< 80 100% > 90% 60% 0–10% 100% 100% > 90% 20–30% 100% > 90% 80–90% 0–10%
30–< 50 100% > 90% 50% 0–10% 100% 100% 90% 40–50% 100% 100% 80–90% 20–30%
< 30 > 90% 50–60% 0–10% 0–10% 100% 100% 80–90% 30–40% 100% > 90% 80–90% 20–30%

Shaded region indicates suboptimal probability of target Css,avg attainment


FDA Food and Drug Administration, EMA European Medicines Agency, CrCl creatinine clearance, Css,avg average steady-state concentration
Curr Infect Dis Rep (2017) 19:38 Page 7 of 9 38

with clinical dose equivalents of polymyxin B [55]. As men- newer beta-lactam/beta-lactamase inhibitor combination
tioned previously, population pharmacokinetic modeling has agents (i.e., ceftolozane/tazobactam and ceftazidime/
suggested that colistin may best be used as part of a highly avibactam), further studies are needed to evaluate the
active combination due to the inability to achieve adequate effects of combining these agents with polymyxins.
plasma concentrations, especially in patients with moderate
to good renal function and/or for organisms with an MIC
≥ 1 mg/L [40]. Conclusion
Given the high rates of colistin resistance in
Enterobacteriaceae and reports of colistin heteroresistance in Polymyxins are one of the last remaining antimicrobial classes
A. baumannii, combination therapy with a polymyxin and an with activity against multi-drug resistant Gram-negative path-
additional agent may be considered in order to avoid develop- ogens. Of great concern is the emergence of polymyxin-resis-
ment of resistance or isolation of resistant subpopulations dur- tance, particularly the plasmid-mediated mcr-1 gene, which
ing therapy. The selection of the antimicrobial agent to be used may be transferable between bacterial species and increasing
in combination with colistin or polymyxin B should be based rates of polymyxin resistance in carbapenem-resistant
upon data regarding synergistic activity of the combination. Enterobacteriaceae (CRE). As our understanding of the
The most common antimicrobials evaluated in combination mechanisms and occurrence of polymyxin resistance grows,
with the polymyxins are the carbapenems. A time-kill study so must our understanding of how to best optimize the use of
of various dosing strategies of polymyxin B in combination these agents including polymyxin agent selection, dosing op-
with doripenem demonstrated rapid and extensive bacterial timization, and use in combination regimens.
killing within 24 h and was sustained over 10 days when
aggressive dosing regimens for polymyxin with doripenem Acknowledgments We thank Elizabeth Neuner, PharmD for assisting
were used [56]. A systematic review of in vitro synergy of with technical editing, language editing, and proofreading of this
manuscript.
polymyxins and carbapenems against Gram-negative bacteria
demonstrated high rates of synergistic activity with the com-
Compliance with Ethical Standards
bination, especially for A. baumannii, as well as less resistance
development [57]. Conflict of Interest The authors declare that they have no conflicts of
The clinical benefits of polymyxin combination ther- interest.
apy are mostly limited to retrospective studies. In a ret-
rospective analysis of colistin monotherapy versus com- Human and Animal Rights This article does not contain any studies
bination therapy in critically ill patients with MDR with human or animal subjects performed by any of the authors.
Gram-negative pneumonia, Parchem and colleagues
demonstrated significant improvements in microbiologi-
cal cure rates with combination therapy; however, no References
difference in clinical cure was observed [58]. Other an-
timicrobial agents that have been evaluated in combina- Papers of particular interest published recently have been
tion with polymyxins include rifampicin and fosfomycin. highlighted as:
Studies comparing colistin monotherapy to a combina- • Of importance
tion of colistin and rifampicin have not demonstrated •• Of major importance
significant microbiological or clinical benefits of the
combination [59, 60]. A preliminary study of colistin 1. U.S. Department of Health and Human Services Centers for
Disease Control and Prevention. Antibiotic resistance threats in
versus a combination of colistin and intravenous
the United States, 2013. 2013 [July 8, 2017]; Available from:
fosfomycin failed to show any clinical benefits from https://www.cdc.gov/drugresistance/pdf/ar-threats-2013-508.pdf.
the combination against carbapenem-resistant 2. Zavascki AP, Goldani LZ, Li J, Nation RL. Polymyxin B for the
A. baumannii infections [61]. As such, the addition of treatment of multidrug-resistant pathogens: a critical review. J
rifampicin or fosfomycin to polymyxin therapy is not Antimicrob Chemother. 2007;60(6):1206–15.
3. Kaye KS, Pogue JM, Tran TB, Nation RL, Li J. Agents of last
routinely recommended. A multicenter randomized con-
resort: polymyxin resistance. Infect Dis Clin N Am. 2016;30(2):
trolled trial comparing colistin monotherapy to colistin 391–414.
plus meropenem for severe infections caused by 4. Cai Y, Chai D, Wang R, Liang B, Bai N. Colistin resistance of
carbapenem-resistant Gram-negative infections is cur- Acinetobacter baumannii: clinical reports, mechanisms and antimi-
rently underway [62]. Based on promising in vitro data, crobial strategies. J Antimicrob Chemother. 2012;67(7):1607–15.
5. Gales AC, Jones RN, Sader HS. Contemporary activity of colistin
we recommend that colistin or polymyxin B be consid- and polymyxin B against a worldwide collection of Gram-negative
ered in combination with highly active bactericidal pathogens: results from the SENTRY Antimicrobial Surveillance
agents such as carbapenems. With the introduction of Program (2006–09). J Antimicrob Chemother. 2011;66(9):2070–4.
38 Page 8 of 9 Curr Infect Dis Rep (2017) 19:38

6.• Sader HS, Farrell DJ, Flamm RK, Jones RN. Antimicrobial suscep- testing methods for colistin in clinical isolates, including
tibility of Gram-negative organisms isolated from patients heteroresistant Enterobacter cloacae and Acinetobacter baumannii
hospitalised with pneumonia in US and European hospitals: results strains. Antimicrob Agents Chemother. 2007;51(10):3726–30.
from the SENTRY Antimicrobial Surveillance Program, 2009– 22. Rodriguez CH, Bombicino K, Granados G, Nastro M, Vay C,
2012. Int J Antimicrob Agents. 2014;43(4):328–34. This study Famiglietti A. Selection of colistin-resistant Acinetobacter
assessed the frequency of colistin resistance in prominent baumannii isolates in postneurosurgical meningitis in an intensive
Gram-negative bacteria isolated from patients hospitalized in care unit with high presence of heteroresistance to colistin. Diagn
US and European medical centers between 2009-2012. Microbiol Infect Dis. 2009;65(2):188–91.
7. Giamarellou H. Epidemiology of infections caused by polymyxin- 23. El-Halfawy OM, Valvano MA. Chemical communication of anti-
resistant pathogens. Int J Antimicrob Agents. 2016;48(6):614–21. biotic resistance by a highly resistant subpopulation of bacterial
8. Walkty A, Lagace-Wiens P, Adam H, Baxter M, Karlowsky J, cells. PLoS One. 2013;8(7):e68874.
Mulvey MR, et al. Antimicrobial susceptibility of 2906 24. Hermes DM, Pormann Pitt C, Lutz L, Teixeira AB, Ribeiro VB,
Pseudomonas aeruginosa clinical isolates obtained from patients Netto B, et al. Evaluation of heteroresistance to polymyxin B
in Canadian hospitals over a period of 8 years: results of the among carbapenem-susceptible and -resistant Pseudomonas
Canadian Ward surveillance study (CANWARD), 2008–2015. aeruginosa. J Med Microbiol. 2013;62(Pt 8):1184–9.
Diagn Microbiol Infect Dis. 2017;87(1):60–3. 25. Landman D, Salamera J, Quale J. Irreproducible and uninterpret-
9. Bradford PA, Kazmierczak KM, Biedenbach DJ, Wise MG, Hackel able Polymyxin B MICs for Enterobacter cloacae and Enterobacter
M, Sahm DF. Correlation of beta-lactamase production and colistin aerogenes. J Clin Microbiol. 2013;51(12):4106–11.
resistance among Enterobacteriaceae isolates from a global surveil- 26. Meletis G, Tzampaz E, Sianou E, Tzavaras I, Sofianou D. Colistin
lance program. Antimicrob Agents Chemother. 2015;60(3):1385– heteroresistance in carbapenemase-producing Klebsiella
92. pneumoniae. J Antimicrob Chemother. 2011;66(4):946–7.
10. Baron S, Hadjadj L, Rolain JM, Olaitan AO. Molecular mecha- 27. Clinical and Laboratory Standards Institute. Performance standards
nisms of polymyxin resistance: knowns and unknowns. Int J for antimicrobial susceptibility testing: twenty-seventh information-
Antimicrob Agents. 2016;48(6):583–91. al supplement. In: Document M100-S27. Wayne, PA: CLSI. 2017.
11. Olaitan AO, Morand S, Rolain JM. Mechanisms of polymyxin 28. The European Committee on Antimicrobial Susceptibility Testing.
resistance: acquired and intrinsic resistance in bacteria. Front Breakpoint tables for interpretation of MICs and zone diameters.
Microbiol. 2014;5:643. Version 7.1, 2017. http://www.eucast.org
12. Yahav D, Farbman L, Leibovici L, Paul M. Colistin: new lessons on 29. Dobrewski R, Savov E, Bernards AT, van den Barselaar M,
an old antibiotic. Clinical Microbiology and Infection: the Official Nordmann P, van den Broek PJ, et al. Genotypic diversity and
Publication of the European Society of Clinical Microbiology and antibiotic susceptibility of Acinetobacter baumannii isolates in a
Infectious Diseases. 2012;18(1):18–29. Bulgarian hospital. Clinical Microbiology and Infection: the
13. Cannatelli A, Giani T, D'Andrea MM, Di Pilato V, Arena F, Conte Official Publication of the European Society of Clinical
V, et al. MgrB inactivation is a common mechanism of colistin Microbiology and Infectious Diseases. 2006;12(11):1135–7.
resistance in KPC-producing Klebsiella pneumoniae of clinical or- 30. Arroyo LA, Garcia-Curiel A, Pachon-Ibanez ME, Llanos AC, Ruiz
igin. Antimicrob Agents Chemother. 2014;58(10):5696–703. M, Pachon J, et al. Reliability of the E-test method for detection of
14. Needham BD, Trent MS. Fortifying the barrier: the impact of lipid colistin resistance in clinical isolates of Acinetobacter baumannii. J
A remodeling on bacterial pathogenesis. Nat Rev Microbiol. Clin Microbiol. 2005;43(2):903–5.
2013;11(7):467–81. 31. European Center for Disease Prevention and Control.
15.•• Liu YY, Wang Y, Walsh TR, Yi LX, Zhang R, Spencer J, et al. Antimicrobial resistance surveillance in Europe 2014. Annual
Emergence of plasmid-mediated colistin resistance mechanism Report of the European Antimicrobial Resistance Surveillance
MCR-1 in animals and human beings in China: a microbiological Network (EARS-Net). Stockholm: ECDC; 2015.
and molecular biological study. Lancet Infect Dis. 2016;16(2):161– 32. Jeannot K, Bolard A, Plesiat P. Resistance to polymyxins in Gram-
8. This study reports the emergence of the first plasmid- negative organisms. Int J Antimicrob Agents. 2017;49(5):526–35.
mediated polymyxin resistance mechanism, mcr-1, in 33. McGann P, Snesrud E, Maybank R, Corey B, Ong AC, Clifford R,
Escherichia coli isolates collected from food animals and et al. Erratum for McGann et al., Escherichia coli harboring mcr-1
highlighted the need for global collaboration in combating and blaCTX-M on a novel IncF plasmid: first report of mcr-1 in the
multidrug-resistant Gram-negative organisms. United States. Antimicrob Agents Chemother. 2016;60(8):5107.
16. Zhi C, Lv L, Yu LF, Doi Y, Liu JH. Dissemination of the mcr-1 34. Centers for Disease Control and Prevention (CDC). Tracking mcr-
colistin resistance gene. Lancet Infect Dis. 2016;16(3):292–3. 1. Available at: https://www.cdc.gov/drugresistance/tracking-mcr1.
17. Castanheira M, Griffin MA, Deshpande LM, Mendes RE, Jones html. [Accessed: 24 July 2017].
RN, Flamm RK. Detection of mcr-1 among Escherichia coli clin- 35. Corbella M, Mariani B, Ferrari C, Comandatore F, Scaltriti E, Marone
ical isolates collected worldwide as part of the SENTRY P, et al. Three cases of mcr-1-positive colistin-resistant Escherichia coli
Antimicrobial Surveillance Program in 2014 and 2015. bloodstream infections in Italy, August 2016 to January 2017. Euro
Antimicrob Agents Chemother. 2016;60(9):5623–4. Surveill. 2017. https://doi.org/10.2807/1560-7917.ES.2017.22.16.
18. El-Halfawy OM, Valvano MA. Antimicrobial heteroresistance: an 30517.
emerging field in need of clarity. Clin Microbiol Rev. 2015;28(1): 36. Elnahriry SS, Khalifa HO, Soliman AM, Ahmed AM, Hussein AM,
191–207. Shimamoto T, et al. Emergence of plasmid-mediated colistin resis-
19. Li J, Rayner CR, Nation RL, Owen RJ, Spelman D, Tan KE, et al. tance gene mcr-1 in a clinical Escherichia coli isolate from Egypt.
Heteroresistance to colistin in multidrug-resistant Acinetobacter Antimicrob Agents Chemother. 2016;60(5):3249–50.
baumannii. Antimicrob Agents Chemother. 2006;50(9):2946–50. 37. Nation RL, Velkov T, Li J. Colistin and polymyxin B: peas in a pod, or
20. Hawley JS, Murray CK, Jorgensen JH. Colistin heteroresistance in chalk and cheese? Clinical infectious diseases: an official publication
Acinetobacter and its association with previous colistin therapy. of the Infectious Diseases Society of America. 2014;59(1):88–94.
Antimicrob Agents Chemother. 2008;52(1):351–2. 38. Bergen PJ, Bulitta JB, Forrest A, Tsuji BT, Li J, Nation RL.
21. Lo-Ten-Foe JR, de Smet AM, Diederen BM, Kluytmans JA, van Pharmacokinetic/pharmacodynamic investigation of colistin
Keulen PH. Comparative evaluation of the VITEK 2, disk diffu- against Pseudomonas aeruginosa using an in vitro model.
sion, etest, broth microdilution, and agar dilution susceptibility Antimicrob Agents Chemother. 2010;54(9):3783–9.
Curr Infect Dis Rep (2017) 19:38 Page 9 of 9 38

39. Cheah SE, Wang J, Nguyen VT, Turnidge JD, Li J, Nation RL. New with serious infections resistant to standard antibiotics. Available at:
pharmacokinetic/pharmacodynamic studies of systemically admin- http://www.ema.europa.eu/docs/en_GB/document_library/
istered colistin against Pseudomonas aeruginosa and Acinetobacter Referrals_document/Polymyxin_31/WC500176333.pdf. Accessed
baumannii in mouse thigh and lung infection models: smaller re- 24 July 2017.
sponse in lung infection. J Antimicrob Chemother. 2015;70(12): 53.•• Nation RL, Garonzik SM, Li J, Thamlikitkul V, Giamarellos-
3291–7. Bourboulis EJ, Paterson DL, et al. Updated US and European dose
40. Garonzik SM, Li J, Thamlikitkul V, Paterson DL, Shoham S, Jacob recommendations for intravenous colistin: how do they perform?
J, et al. Population pharmacokinetics of colistin methanesulfonate Clinical Infectious Diseases: an Official Publication of the
and formed colistin in critically ill patients from a multicenter study Infectious Diseases Society of America. 2016;62(5):552–8. This
provide dosing suggestions for various categories of patients. study was a population pharmacokinetic analysis comparing
Antimicrob Agents Chemother. 2011;55(7):3284–94. US FDA versus EMA versus population pharmacokinetics
41. Horcajada JP, Sorli L, Luque S, Benito N, Segura C, Campillo N, based dosing recommendations for colistin that found funda-
et al. Validation of a colistin plasma concentration breakpoint as a mental differences in the probability of target steady state con-
predictor of nephrotoxicity in patients treated with colistin centration attainment between US FDA and EMA dosing rec-
methanesulfonate. Int J Antimicrob Agents. 2016;48(6):725–7. ommendations, especially in patients with low body weight and
42.•• Nation RL, Garonzik SM, Thamlikitkul V, Giamarellos-Bourboulis impaired renal function.
EJ, Forrest A, Paterson DL, et al. Dosing guidance for intravenous 54. Sandri AM, Landersdorfer CB, Jacob J, Boniatti MM, Dalarosa
colistin in critically-ill patients. Clinical infectious diseases: an of- MG, Falci DR, et al. Population pharmacokinetics of intravenous
ficial publication of the Infectious Diseases Society of America. polymyxin B in critically ill patients: implications for selection of
2017;64(5):565–71. This study was a multinational population dosage regimens. Clinical Infectious Diseases: an Official
pharmacokinetic analysis of various colistin dosing strategies Publication of the Infectious Diseases Society of America.
that aimed to derive a dosing algorithm for achieving desired 2013;57(4):524–31.
steady state plasma concentrations of colistin based on specific 55. Tam VH, Schilling AN, Vo G, Kabbara S, Kwa AL, Wiederhold
patient characteristics. NP, et al. Pharmacodynamics of polymyxin B against
43. Pogue JM, Ortwine JK, Kaye KS. Clinical considerations for opti- Pseudomonas aeruginosa. Antimicrob Agents Chemother.
mal use of the polymyxins: a focus on agent selection and dosing. 2005;49(9):3624–30.
Clinical Microbiology and Infection: the Official Publication of the 56. Rao GG, Ly NS, Bulitta JB, Soon RL, San Roman MD, Holden PN,
European Society of Clinical Microbiology and Infectious et al. Polymyxin B in combination with doripenem against
Diseases. 2017;23(4):229–33. heteroresistant Acinetobacter baumannii: pharmacodynamics of
44. Oliveira MS, Prado GV, Costa SF, Grinbaum RS, Levin AS. new dosing strategies. J Antimicrob Chemother. 2016;71(11):
Polymyxin B and colistimethate are comparable as to efficacy 3148–56.
and renal toxicity. Diagn Microbiol Infect Dis. 2009;65(4):431–4.
57. Zusman O, Avni T, Leibovici L, Adler A, Friberg L, Stergiopoulou
45. Crass RL, Rutter WC, Burgess DR, Martin CA, Burgess DS.
T, et al. Systematic review and meta-analysis of in vitro synergy of
Nephrotoxicity in patients with or without cystic fibrosis treated
polymyxins and carbapenems. Antimicrob Agents Chemother.
with polymyxin B compared to colistin. Antimicrob Agents
2013;57(10):5104–11.
Chemother. 2017. https://doi.org/10.1128/AAC.02329-16.
46. Phe K, Lee Y, McDaneld PM, Prasad N, Yin T, Figueroa DA, et al. 58. Parchem NL, Bauer KA, Cook CH, Mangino JE, Jones CD, Porter
In vitro assessment and multicenter cohort study of comparative K, et al. Colistin combination therapy improves microbiologic cure
nephrotoxicity rates associated with colistimethate versus polymyx- in critically ill patients with multi-drug resistant gram-negative
in B therapy. Antimicrob Agents Chemother. 2014;58(5):2740–6. pneumonia. European Journal of Clinical Microbiology &
47. Rigatto MH, Oliveira MS, Perdigao-Neto LV, Levin AS, Carrilho Infectious Diseases: Official Publication of the European Society
CM, Tanita MT, et al. Multicenter prospective cohort study of renal of Clinical Microbiology. 2016;35(9):1433–9.
failure in patients treated with colistin versus polymyxin B. 59. Aydemir H, Akduman D, Piskin N, Comert F, Horuz E, Terzi A,
Antimicrob Agents Chemother. 2016;60(4):2443–9. et al. Colistin vs. the combination of colistin and rifampicin for the
48. Tuon FF, Rigatto MH, Lopes CK, Kamei LK, Rocha JL, Zavascki treatment of carbapenem-resistant Acinetobacter baumannii
AP. Risk factors for acute kidney injury in patients treated with ventilator-associated pneumonia. Epidemiol Infect. 2013;141(6):
polymyxin B or colistin methanesulfonate sodium. Int J 1214–22.
Antimicrob Agents. 2014;43(4):349–52. 60. Durante-Mangoni E, Signoriello G, Andini R, Mattei A, De
49. Akajagbor DS, Wilson SL, Shere-Wolfe KD, Dakum P, Charurat Cristoforo M, Murino P, et al. Colistin and rifampicin compared
ME, Gilliam BL. Higher incidence of acute kidney injury with with colistin alone for the treatment of serious infections due to
intravenous colistimethate sodium compared with polymyxin B in extensively drug-resistant Acinetobacter baumannii: a multicenter,
critically ill patients at a tertiary care medical center. Clinical randomized clinical trial. Clinical Infectious Diseases: an Official
Infectious Diseases: an Official Publication of the Infectious Publication of the Infectious Diseases Society of America.
Diseases Society of America. 2013;57(9):1300–3. 2013;57(3):349–58.
50. Falagas ME, Kasiakou SK, Tsiodras S, Michalopoulos A. The use 61. Sirijatuphat R, Thamlikitkul V. Preliminary study of colistin versus
of intravenous and aerosolized polymyxins for the treatment of colistin plus fosfomycin for treatment of carbapenem-resistant
infections in critically ill patients: a review of the recent literature. Acinetobacter baumannii infections. Antimicrob Agents
Clin Med Res. 2006;4(2):138–46. Chemother. 2014;58(9):5598–601.
51. FDA Approved Drug Products. Label and approval history for 62. Dickstein Y, Leibovici L, Yahav D, Eliakim-Raz N, Daikos GL,
Coly-Mycin M, NDA 050108. Available at: http://www. Skiada A, et al. Multicentre open-label randomised controlled trial
accessdata.fda.gov/drugsatfda_docs/label/2013/050108s030lbl. to compare colistin alone with colistin plus meropenem for the
pdf. Accessed 24 July 2017. treatment of severe infections caused by carbapenem-resistant
52. European Medicines Agency completes review of polymyxin- Gram-negative infections (AIDA): a study protocol. BMJ Open.
based medicines: recommendations issued for safe use in patients 2016;6(4):e009956.

You might also like