ArticlePDF AvailableLiterature Review

The use of combination therapy for the improvement of colistin activity against bacterial biofilm

Authors:
  • College of Medicine, University of Thi-Qar, Al-Nasiriyah,Iraq.

Abstract

Colistin is used as a last resort for the management of infections caused by multi-drug resistant (MDR) bacteria. However, the use of this antibiotic could lead to different side effects, such as nephrotoxicity, in most patients, and the high prevalence of colistin-resistant strains restricts the use of colistin in the clinical setting. Additionally, colistin could induce resistance through the increased formation of biofilm; biofilm-embedded cells are highly resistant to antibiotics, and as with other antibiotics, colistin is impaired by bacteria in the biofilm community. In this regard, the researchers used combination therapy for the enhancement of colistin activity against bacterial biofilm, especially MDR bacteria. Different antibacterial agents, such as antimicrobial peptides, bacteriophages, natural compounds, antibiotics from different families, N-acetylcysteine, and quorum-sensing inhibitors, showed promising results when combined with colistin. Additionally, the use of different drug platforms could also boost the efficacy of this antibiotic against biofilm. The mentioned colistin-based combination therapy not only could suppress the formation of biofilm but also could destroy the established biofilm. These kinds of treatments also avoided the emergence of colistin-resistant subpopulations, reduced the required dosage of colistin for inhibition of biofilm, and finally enhanced the dosage of this antibiotic at the site of infection. However, the exact interaction of colistin with other antibacterial agents has not been elucidated yet; therefore, further studies are required to identify the precise mechanism underlying the efficient removal of biofilms by colistin-based combination therapy.
Vol.:(0123456789)
1 3
Brazilian Journal of Microbiology
https://doi.org/10.1007/s42770-023-01189-7
CLINICAL MICROBIOLOGY - REVIEW
The use ofcombination therapy fortheimprovement ofcolistin
activity againstbacterial biofilm
AbduladheemTurkiJalil1 · RawaaTurkiAbdulghafoorAlrawe2· MontahaA.Al‑Saar3·
MurtadhaLaftaShaghnab4· MunaS.Merza5· MuntherAbosaooda6· RahimLatef7
Received: 14 July 2023 / Accepted: 27 October 2023
© The Author(s) under exclusive licence to Sociedade Brasileira de Microbiologia 2023
Abstract
Colistin is used as a last resort for the management of infections caused by multi-drug resistant (MDR) bacteria. However,
the use of this antibiotic could lead to different side effects, such as nephrotoxicity, in most patients, and the high prevalence
of colistin-resistant strains restricts the use of colistin in the clinical setting. Additionally, colistin could induce resistance
through the increased formation of biofilm; biofilm-embedded cells are highly resistant to antibiotics, and as with other anti-
biotics, colistin is impaired by bacteria in the biofilm community. In this regard, the researchers used combination therapy
for the enhancement of colistin activity against bacterial biofilm, especially MDR bacteria. Different antibacterial agents,
such as antimicrobial peptides, bacteriophages, natural compounds, antibiotics from different families, N-acetylcysteine, and
quorum-sensing inhibitors, showed promising results when combined with colistin. Additionally, the use of different drug
platforms could also boost the efficacy of this antibiotic against biofilm. The mentioned colistin-based combination therapy
not only could suppress the formation of biofilm but also could destroy the established biofilm. These kinds of treatments
also avoided the emergence of colistin-resistant subpopulations, reduced the required dosage of colistin for inhibition of
biofilm, and finally enhanced the dosage of this antibiotic at the site of infection. However, the exact interaction of colistin
with other antibacterial agents has not been elucidated yet; therefore, further studies are required to identify the precise
mechanism underlying the efficient removal of biofilms by colistin-based combination therapy.
Keywords Biofilm· Combination therapy· Colistin
Introduction
Colistin is an antimicrobial agent that is extracted from Pae-
nibacillus polymyxa, which belongs to class E of the poly-
mixin group [1]. Two forms of colistin sulfate and colistin
methane sulfonate are used in oral-topical and injectable
forms for humans, respectively [2]. The general mechanism
and goals of colistin are binding to the negative charge of
lipopolysaccharides (LPS) in the outer membrane of Gram-
negative bacteria, increasing membrane permeability and
bacterial lysis, producing reactive oxygen species, and inhib-
iting respiratory enzymes [3]. In addition to the fact that
some bacteria, such as Proteus species, Morganella mor-
ganii, Serratia species, Providencia species, Burkholderia
pseudomallei, Neisseria species, and Edwardsiella tarda,
are intrinsically resistant to colistin, resistance can be seen
through chromosomal mutations of genes involved in LPS
biosynthesis or glucose transport pathways and the transfer
of the mcr-1 gene through a plasmid [4].
Responsible Editor: Beatriz Ernestina Cabilio Guth
* Abduladheem Turki Jalil
abedalazeem799@gmail.com
1 College ofMedicine, University ofThi-Qar, Al-Nasiriya,
Iraq
2 Education College forWomen, Department ofBiology,
University ofAnbar, Anbar, Iraq
3 Community Health Department, Institute ofMedical
Technology/Baghdad, Middle Technical University,
Baghdad, Iraq
4 National University ofScience andTechnology, Nasiriyah,
DhiQar, Iraq
5 Prosthetic Dental Techniques Department, Al-Mustaqbal
University College, Babylon51001, Iraq
6 Medical Laboratory Technology Department, College
ofMedical Technology, The Islamic University, Najaf, Iraq
7 Medical Technical College, Al-Farahidi University, Baghdad,
Iraq
Brazilian Journal of Microbiology
1 3
Colistin is often used as a last line of defense in critical
clinical conditions such as bacteremia/sepsis and ventilator-
associated pneumonia in the intensive care unit and against
Gram-negative bacteria such as Pseudomonas spp., Acine-
tobacter spp., and Enterobacteriales [5]. According to the
studies, the prevalence of resistance to colistin is increasing,
especially in multi-drugs resistant (MDR) bacteria, and the
scientific community has demanded to reduce the use of
colistin [1]. In addition, one of the limitations of the wide-
spread use of colistin is the high incidence of poisoning,
such as renal and neurotoxicity, neuromuscular blockade,
and, in some cases, fatal manifestations [6].
The point to consider is that one of the biggest problems
in the treatment of bacterial infections is the formation of
biofilm by the most important pathogens, and the activity of
colistin is often impaired when faced with biofilm [7]. Bio-
film formation is a bacterial community in which microor-
ganisms are enclosed in an exopolysaccharide matrix, which
leads to extensive phenotypic changes in the bacterial popu-
lation and makes them 1–1000 times more resistant than
their planktonic form [7, 8].
Noteworthy, a recently published study reported that a
minimum inhibitory concentration (MIC)-dependent con-
centration of colistin could not suppress Escherichia coli
growth. On the other hand, this antibiotic could lead to bac-
terial regrowth, with the possibility of resulting in colistin
resistance. Moreover, increased expression of the phoQ gene
could lead to increased biofilm formation in colistin-induced
resistance bacteria, which prevents colistin from reaching
the site of action, finally inducing antibiotic resistance [9].
Therefore, in addition to the increased prevalence of colistin-
resistant strains in recent years, the biofilm community could
also decrease the inhibitory activity of this antibiotic.
To this end, researchers have used new therapeutic
options, including combination therapy, to increase the
activity of colistin against bacterial biofilm, especially
MDR isolates [10]. Antimicrobial peptides, bacteriophages,
natural compounds and natural products, antibiotics, N-ace-
tylcysteine, and quorum-sensing inhibitors are among the
agents that have shown promising results when used with
colistin. The purpose of combined treatment is to overcome
resistance to colistin and other antimicrobial agents, increase
the effectiveness of the combination compared to mono-
therapy, and reduce the toxicity associated with colistin [6,
11]. It should be noted that previous studies have shown the
existence of synergistic interactions when combining colistin
with antimicrobial drugs against Gram-negative clinical iso-
lates and reducing hospital mortality [12]. Considering the
therapeutic and practical role of colistin in the clinic and as
the last line of treatment, the purpose of this review study is
to examine and discuss the combined use of this drug along
with other therapeutic agents and drug carriers for inhibition
and destruction of bacterial biofilm in a comprehensive way.
Biolm
Bacterial organisms have been found to undergo evolu-
tionary processes in response to various obstacles and dif-
ficulties encountered in hostile settings. These problems
may encompass those that are induced by the existence
of host immunity, the presence of an antimicrobial agent,
and restrictions in nutritional availability. Biofilm pro-
duction is a notable survival strategy employed by bac-
teria [1315]. A biofilm refers to a complex assemblage
of many microbial organisms that are enveloped inside
a self-generated polymeric matrix. This matrix serves to
anchor the biofilm to either living (biotic) or non-living
(abiotic) surfaces. It is worth noting that the production
of biofilms is a capability exhibited by nearly all bacteria,
given the presence of appropriate environmental condi-
tions [16]. Bacterial aggregation and biofilm maturation
encompass both reversible and irreversible stages, which
are influenced by numerous conserved and/or species-spe-
cific variables. During the initial stage, the microorganism
establishes a reversible attachment to a surface by engag-
ing in weak contact, such as van der Waals forces, with
either an abiotic or biotic surface. Various surfaces can
exist in different forms, encompassing earth and aquatic
systems as well as indwelling medical equipment. Addi-
tionally, surfaces can be found in living tissues, such as
heart valves, tooth enamel, the lung, and the middle ear
[17]. The second stage involves the process of irreversible
attachment, which is facilitated by flagella, pili, and other
surface appendages, as well as specific receptors [18].
The accumulation of multilayered cells occurs through
cellular division; after which they initiate the synthesis of
their own extracellular polymeric substance (EPS) matrix.
This matrix mostly consists of polysaccharides, proteins,
and extracellular DNA, which play a crucial role in pro-
moting the formation of the biofilm [19]. The detection of
EPSs can be achieved through microscopic examination as
well as chemical analysis. EPSs serve as the foundational
matrix or structural framework for the formation and main-
tenance of biofilms. EPS has a high level of hydration,
with approximately 98% of its composition consisting of
water. Additionally, EPS displays a strong adherence to the
underlying surface. Furthermore, the EPS biofilm matrix
serves as a protective barrier for microbes, shielding them
from the effects of antimicrobial medications and the host
immune system. Hence, the microorganisms implicated in
co-infections have the ability to form polymicrobial bio-
films, which exhibit not only inherent genotypic resistance
but also phenotypic resistance or tolerance to antimicrobial
medicines that are linked to the biofilm matrix [20]. Water
channels are present in a fully developed biofilm, facili-
tating the efficient distribution of nutrients and signaling
Brazilian Journal of Microbiology
1 3
chemicals throughout the biofilm structure. Biofilm cells
exhibit detachment either individually or in aggregates due
to both internal and extrinsic stimuli. Ultimately, these
cells become dispersed, leading to their colonization of
various ecological niches [13].
Biofilm-associated organisms have a reduced growth rate
compared to planktonic organisms, perhaps because of the
constraints imposed by nutrition and/or oxygen deprivation
on the cells [21]. Cell detachment from the biofilm occurs
due to two primary mechanisms: cellular proliferation
and division, or the elimination of biofilm aggregates that
encompass substantial quantities of cells. The potential for
detached cells to induce a systemic infection is contingent
upon various aspects, encompassing the host immune sys-
tem’s reaction [22]. Biofilms hold considerable importance
in the field of public health due to the fact that microor-
ganisms associated with biofilms demonstrate a significant
reduction in their susceptibility to antimicrobial treatments.
The biofilm represents a suitable niche for the exchange
of resistance genes [17, 23]. The impact on susceptibil-
ity can be categorized as either intrinsic, which refers to
characteristics inherent in the biofilm mode of growth, or
acquired, which is associated with the acquisition of resist-
ance plasmids. There exist a minimum of three rationales
accounting for the inherent antimicrobial resistance exhib-
ited by biofilms. Initially, it is imperative for antimicrobial
drugs to effectively permeate the EPS matrix in order to
establish contact with and neutralize the microorganisms
residing within the biofilm [24]. EPSs inhibit the diffusion
process through two mechanisms: chemical reactions with
antimicrobial compounds and restriction of their transport
rate. Additionally, organisms linked with biofilms exhibit
diminished growth rates, resulting in a decreased uptake of
antimicrobial drugs into the cell. Consequently, this phe-
nomenon impacts the kinetics of inactivation [25]. Further-
more, the local environment encompassing the cells inside a
biofilm may offer favorable conditions that serve to enhance
the organism’s protection [25]. In relation to acquired resist-
ance, studies have demonstrated that the exchange of plas-
mids within biofilms can occur under many circumstances.
Numerous bacterial species have demonstrated the ability
to transfer plasmids to diverse bacterial counterparts. The
potential factors contributing to increased plasmid transfer
within biofilms encompass the heightened likelihood of cel-
lular interaction and the little impact of shear pressures on
both the disruption of cell-to-cell contact and the integrity
of the pili essential for conjugation [26].
Finally, it is noteworthy to mention that a crucial aspect of
addressing biofilm is the recognition of polymicrobial bio-
film communities that engage in mutual interactions, result-
ing in a synergistic impact. These interactions frequently
result in an elevated level of resistance to both host and
antimicrobial drugs across all participating species [27, 28].
This phenomenon can arise from either heightened tolerance
or adaptive resistance, which stem from the synergistic inter-
actions between different species, or from the presence of
antibiotic-resistant organisms inside a polymicrobial biofilm,
which confers protection to other species within the biofilm
against antibiotic interventions. Species inside polymicro-
bial biofilms frequently exhibit heightened pathogenicity, an
enhanced capacity to digest and exploit organic molecules
within their surroundings, and provide a conducive milieu
for the dissemination of adaptive characteristics and genes
associated with antimicrobial resistance, both within and
across species [29, 30]. Due to this feature, the management
of diseases associated with microbial biofilms has emerged
as a highly complex issue within the healthcare system. The
cumulative impact of infections linked to biofilms can be
highly debilitating for patients, as these illnesses have the
ability to persist for extended periods, leading to a loss of
hope for patients regarding their recovery. In particular, bio-
film has been identified in various types of wounds, includ-
ing chronic leg ulcers, diabetic foot ulcers, pressure ulcers,
burns, malignant wounds, and surgical wounds [3133].
Combination use ofcolistin withother
antibiotics
Researchers are considering combining colistin with other
antibiotics to improve biofilm eradication. Noteworthy, anti-
biotics from different families inhibit bacteria using various
mechanisms, and synergy assessment has clarified the inter-
action of two drugs in combination against bacterial isolates.
Therefore, combination therapy could suppress bacterial
communities more efficiently in comparison to monotherapy.
To this end, researchers considered colistin-carbapenems
combination therapy to inhibit the biofilm community of
Gram-negative bacteria. In a recently published study, dif-
ferent concentrations of colistin-meropenem showed a syn-
ergistic effect against the biofilm structures of A. baumannii
and P. aeruginosa strains, while an indifference effect was
found against K. pneumoniae [34]. The results of another
investigation also showed that colistin-meropenem combi-
nation therapy decreases the biofilm formation of Myroides
odoratimimus strains by 92.4%. Noteworthy, colistin showed
a low inhibitory effect against biofilm when used alone,
while the level of inhibition improved approximately three-
fold when used in combination with meropenem or cip-
rofloxacin [35]. In line with these findings, other studies
also reported the synergistic effect of colistin-carbapenems
combination therapy against a biofilm community of differ-
ent bacteria [3638]. In this regard, Tamayo etal. reported
that the colistin-doripenem combination not only showed
synergistic effects against biofilm-embedded P. aeruginosa
cells but also reduced the emergence of colistin resistance
Brazilian Journal of Microbiology
1 3
strains. The authors proposed that colistin’s ability to destroy
the outer membrane of Gram-negative bacteria enhances
the permeability of these bacteria and could allow greater
access of doripenem to the critical penicillin-binding pro-
teins located on the cytoplasmic membrane, where the car-
bapenems act [38].
On the other hand, the results of a study published in
2019 indicated that the addition of colistin to meropenem
produced no relevant benefits against extended-spectrum-
β-lactamase (ESBL)-producing K. pneumoniae; however,
this combination therapy protected against the emergence
of colistin-resistant subpopulations [39]. Other beta-lac-
tams, such as ceftazidime, also showed a synergistic effect
in combination with colistin against bacterial biofilm. In this
regard, a recently published study reported that colistin mon-
otherapy could significantly destroy the biofilm structure of
P. aeruginosa, but monotherapy leads to the emergence of
colistin-resistant strains. Nonetheless, colistin-ceftazidime
combination therapy prevented resistance emergence to
both antibiotics and improved killing in comparison to each
monotherapy [40]. This supports the finding by Gómez-
Junyent etal., who reported colistin plus meropenem and
ceftolozane/tazobactam as the most applicable therapeutic
approaches for the treatment of MDR and extensively drug-
resistant (XDR) P. aeruginosa biofilm-associated infections,
respectively [41]. Although the interaction of colistin and
beta-lactams against biofilm structure is not well known,
the findings of previous studies have shown that bacteria
with low metabolic profiles within the biofilms are more
susceptible to colistin, while bacteria that are more meta-
bolically active and present in the outer layers of the biofilm
are more susceptible to beta-lactams [40, 4245]. Therefore,
colistin/beta-lactam combination therapy can efficiently kill
both groups of bacteria and reduce the emergence of antibi-
otic resistance. Additionally, as mentioned, colistin destroys
the outer structure of biofilm and facilitates the penetration
of beta-lactams to subpopulations within biofilm layers.
Altogether, colistin and beta-lactam’s functions on various
biofilm layers and cellular pathways could explain the syn-
ergy observed with the combination. Furthermore, colistin/
beta-lactam combination therapy could effectively destroy
the biofilm community of MDR bacteria and reduce the
emergence of colistin-resistant strains. However, it seems
this combination therapy has variable effects against vari-
ous Gram-negative bacteria; thus, further studies are needed
to evaluate whether the benefits of the colistin/beta-lactam
combination therapy are obtained over longer periods or
against different carbapenem-resistant or ESBL-producing
bacteria.
In addition to beta-lactams, rifampicin, also in combina-
tion with colistin, showed promising results for the inhibi-
tion of bacterial biofilm [37, 46, 47]. Geladari etal. reported
that colistin plus rifampicin interacted in synergy to decrease
the viability of carbapenem-resistant K. pneumoniae biofilm
cells at low rifampicin concentrations. Notably, the syner-
gies observed with this combination therapy were higher
than those observed with other combinations such as colis-
tin/meropenem and colistin/tigecycline [37]. Of note, the
authors supposed that colistin leads to the disruption of the
outer bacterial membrane and improves the penetration and
intracellular concentration of rifampicin to suppress DNA
transcription or the mutual killing of resistant subpopula-
tions by each drug [37, 48, 49]. Additionally, rifampicin’s
ability to penetrate biofilm cells is supported by clinical
combination studies using rifampicin to treat prosthetic
material infections [49]. The result of another study demon-
strated that polymixin-rifampicin combination therapy, after
one hour, reduced the gene expression of quorum-sensing
(QS)-regulated virulence factors, such as biofilm formation
and secretion systems of P. aeruginosa. On the other hand,
this combination therapy, after four hours, enhanced the
expression of peptidoglycan biosynthesis genes. Notably,
the combination therapy caused a substantial accumulation
of nucleotides and amino acids that lasted at least 4h, indi-
cating that bacterial cells were in a state of metabolic arrest
[46]. Therefore, the colistin-rifampicin combination therapy
had a synergistic effect against the biofilm community of
different bacteria at relatively low concentrations, and this
combination therapy should be considered for the manage-
ment of biofilm-associated infection and warrant further
assessment in appropriate invivo models.
In the end, the double combination of colistin and tigecy-
cline also showed promising inhibitory effects against bac-
terial biofilm. Tigecycline, a member of the glycylcycline
class of semisynthetic antimicrobial agents, has the potential
for disruption of biofilm structure; however, the findings of
the invitro catheter model study showed that tigecycline
monotherapy could be related to the regrowth of bacteria
[50]. To this end, the combination of this antibiotic with
colistin was considered by researchers for better inhibition of
biofilm cells. Sato etal. reported that the biofilm community
of MDR-A. baumannii was eradicated with colistin but not
tigecycline. Noteworthy, the combination usage of colistin
with a high concentration of tigecycline effectively eradi-
cated biofilm, while attenuation happened with the com-
bination of colistin and low concentrations of tigecycline
[51]. In line with these results, another study also reported
that just high concentrations of these drugs could lead to
synergistic effects in the double combination therapy against
K. pneumonia biofilm, whereas at low concentrations, the
combination treatment indicated indifferent results [37]. It
is noteworthy to mention that efflux pumps are considered
important factors for virulence and antibiotic resistance
in Gram-negative bacteria [52]. Previous studies reported
that different efflux pumps such as EmrAB have an impor-
tant role in resistance to colistin, and low concentrations
Brazilian Journal of Microbiology
1 3
of tigecycline lead to the upregulation of this efflux pump
[53, 54]. Therefore, the authors suggested that one possible
explanation for this observation might be that a low con-
centration of tigecycline increases the expression of efflux
pumps and consequently attenuates the bactericidal activity
of colistin in combination therapy [51].
Collectively, in combination therapy, the second antibi-
otic targeting distinct bacterial subpopulations with different
antimicrobial susceptibilities could complete the function
of colistin against bacterial biofilm. In addition to this so-
called subpopulation synergy (where different drugs target
cells with different susceptibilities), mechanistic synergy
has also been proposed for combinations involving colistin,
whereby each drug acts on different metabolic pathways or
otherwise enhances killing by the second drug [38]. How-
ever, the mechanism involved in the synergistic efficacy of
the combination of colistin and other antibiotics is not well
known; therefore, further studies using validated invitro and
animal biofilm models are needed.
Finally, it is worth noting that other antibiotics such as
clarithromycin, levofloxacin, azithromycin, fosfomycin,
minocycline, mefloquine, and aminoglycosides were also
used in combination with colistin for the inhibition of bac-
terial biofilms (Table1).
Drug delivery platforms
Colistin, as mentioned, is considered a promising therapeutic
approach for the management of MDR Gram-negative bac-
teria. However, the increased prevalence of colistin-resistant
strains in recent years has restricted the clinical usage of
this antibiotic [84]. Noteworthy, recently published stud-
ies reported that different nanoparticles and drug platforms
could be used for different clinical purposes [8587]. To
this end, the use of various drug platforms is suggested by
researchers to deliver intact colistin at the site of infection
and shield its interactions with bacterial biofilm and airway
mucus, thereby enhancing the interaction of this antibiotic
with bacteria.
Sans-Serramitjana etal. proposed that nanoencapsula-
tion could improve the efficacy of colistin against MDR
infections by overcoming the limitations of conventional
pharmaceutical forms. In this concept, these authors evalu-
ated the antibiofilm activity of nanostructured lipid carriers
(NLC)—colistin and free colistin against P. aeruginosa. The
findings indicated the more rapid killing of P. aeruginosa
bacterial biofilms by NLC-colistin than by free colistin [88].
In line with these results, another study that was pub-
lished in 2016 also reported that NLC-colistin had the
same antibacterial function as free colistin against the
planktonic community of P. aeruginosa; nonetheless,
nanoencapsulated colistin was much more efficient in the
eradication of biofilms than free colistin [89]. Further-
more, nano-embedded microparticles (NEMs) for sus-
tained delivery of colistin in the lung were used in another
investigation. To this purpose, the emulsion/solvent diffu-
sion technique was used for the production of poly(lactide-
co-glycolide) (PLGA) containing colistin. Noteworthy,
poly (vinyl alcohol) (PVA) and chitosan (CS) were used
to modulate surface properties and enhance the transport
of synthesized nanoparticles through artificial cystic fibro-
sis (CF) mucus. Additionally, these nanoparticles were
spray-dried in various carriers for the production of MEM.
Colistin-loaded NEM significantly removed the biofilm of
P. aeruginosa and showed a prolonged efficacy in biofilm
eradication compared to the free drug. The results of the
confocal analysis confirmed that the antibiofilm activity
of nanoparticles could be associated with their ability to
penetrate into bacterial biofilms and to sustain the release
of colistin inside the biofilm community [90].
Notably, the coating of the nanoparticle surfaces with chi-
tosan could facilitate the transport of nanoparticles through
mucus, probably as a consequence of mucus fiber collapse,
and produce large channels that can paradoxically enhance
the penetration of cationic chitosan-modified nanoparticles.
Therefore, the engineering of PLGA nanoparticles contain-
ing colistin should be considered for killing bacterial bio-
film, especially in patients with CF [91]. Taken together, the
possible entrapment and slow penetration of colistin within
the biofilm due to electrostatic interactions with the nega-
tively charged alginate matrix can significantly decrease the
availability of colistin in the bottom layers of the biofilm. In
this regard, the use of different nanoplatforms such as NLC
and PLGA could easily penetrate the biofilm structure; thus,
the colistin-nanoplatform can reach the bacteria located in
the deeper layers of the biofilm faster and more easily than
free colistin [88, 92, 93].
Osteomyelitis, one of the most important local infec-
tions, is mostly caused by Methicillin-resistant Staphylo-
coccus aureus (MRSA); nevertheless, the prevalence of
Gram-negative associated bone infections has significantly
increased over the last few years [94]. To this end, different
bacteria such as E. coli, P. aeruginosa, and A. baumannii
have attracted much attention owing to their ability to reach
antibiotic multi-resistance. Additionally, bacterial biofilm
leads to antibiotic resistance and is responsible for prolonged
antibiotic treatment and an aggressive surgical approach in
osteomyelitis [95, 96]. As mentioned in the previous parts,
the reports of colistin-resistant bacteria were increased in
recent years, and this drug is not available in bone void fill-
ers for local high-dose delivery [97]. In this regard, the use
of different drug platforms was considered by researchers
for the enhancement of colistin’s efficacy against bacterial
biofilm and the improvement of the concentration of this
antibiotic at the site of infection.
Brazilian Journal of Microbiology
1 3
Table 1 Other studies used colistin-based combination therapy for the inhibition of bacterial biofilm
Antibacterial agents Class Bacterium Methods Outcome and comments Reference
OligoG CF-5/20 7Alginate oligomer P. aeruginosa High-molecular-weight alginate polymer
bead and mouse lung infection model
The combination therapy remarkably
decreased the MBEC for colistin.
[55]
Low-molecular-weight alginate oligosac-
charide
Alginate oligosaccharide P. aeruginosa CLSM and SEM evaluation Combining therapy effectively destroyed
both intercolony branching/bridging
and microcolony structures in both non-
mucoid and mucoid models.
[56]
Alginate oligosaccharide Alginate oligosaccharide P. aeruginosa Greiner glass-bottomed optical 96-well
plate, CLSM
This compound significantly induced
bacterial death and reduced the formation
of biofilm.
[57]
D-amino acids Amino acid P. aeruginosa CLSM, 96-well plates and biofilm dispersal
assays
The addition of D-amino acids improved
the colistin function and reduced the
count of viable bacteria and MBIC.
[58]
Clarithromycin Antibiotic A. baumannii In vitro antibiotic lock model Colistin-clarithromycin combination
therapy showed bactericidal activity
against bacteria embedded in the biofilm
community.
[59]
Amikacin and levofloxacin Antibiotic P. aeruginosa (CR-isolates) MBECs were examined by counting the
live bacteria in the biofilm, CLSM, and
animal biofilm infection model.
Combined use of colistin with levofloxacin
or amikacin should be considered for
inhibition of biofilm-associated infec-
tions.
[60]
Azithromycin Antibiotic K. pneumonia Biofilm formation in vials and polystyrene
96-well plates
Azithromycin can improve the effectiveness
of colistin.2
[61]
Fosfomycin Antibiotic Different GNB Biofilm chequerboard and quantitative
antibiofilm assays
Colistin-fosfomycin combination therapy
showed a synergistic effect against the
biofilm community of the majority of
tested strains.
[62]
Fosfomycin Antibiotic CR P. aeruginosa Biofilm formation in polystyrene 96-well
plates
Colistin-fosfomycin combination therapy
showed an inhibitory effect against
biofilm.
[36]
Ceftazidime-avibactam Antibiotic XDR P. aeruginosa MTT Methythiazolyl tetrazolium assay The combination therapy can suppress
the biofilm formation and decrease the
production of drug-resistant bacteria.
[63]
Clarithromycin or esomeprazole Antibiotic K. pneumonia In vitro catheter biofilm model The combination therapy showed a syner-
gistic effect.
[50]
Rifampicin Antibiotic CA A. baumannii Polystyrene microtiter plate biofilm assay Monotherapy was not effective at reducing
bacteria in biofilm, while colistin-
rifampicin combination therapy signifi-
cantly decreased the bacteria.
[47]
Clarithromycin Antibiotic P. aeruginosa In vitro catheter biofilm model The combination therapy was most effec-
tive at reducing bacterial count in biofilm
in comparison to the monotherapy.
[64]
Tobramycin Antibiotic P. aeruginosa Static and dynamic biofilm experiments.
CLSM analysis and lung infection in the
animal model
The antibiotic combination significantly
decreased the bacterial count and was
more effective in managing infection in
the animal model than monotherapy.
[65]
Inhaled combination dry powder formula-
tion of colistin and rifapentine
Antibiotic and drug delivery P. aeruginosa Polystyrene microtiter plate biofilm assay Combination dry powder increased the anti-
bacterial against the biofilm community.
[66]
Mefloquine Antimalarial medicine P. aeruginosa Biofilm eradication test and biofilm forma-
tion inhibition test
The combination therapy decreased biofilm
formation and removed pre-formed
established biofilms.
[67]
Brazilian Journal of Microbiology
1 3
Table 1 (continued)
Antibacterial agents Class Bacterium Methods Outcome and comments Reference
Nisin Lantibiotics; bacteriocins P. aeruginosa Static microtiter plate assays Nisin could significantly reduce the
concentration of colistin for inhibition
of biofilm.
[68]
Enterocin DD14 and nisin Bacteriocins Colistin-resistant E.coli Microtiter plate biofilm assay The combination therapy eradicated bacte-
rial biofilm
[69]
Chitosan-coated human albumin nanopar-
ticles
Drug delivery A. baumannii, K. pneumonia Biofilm formation in polystyrene 96-well
plates
This compound inhibited the formation of
biofilm 4- and 60-fold higher than free
colistin.
[3]
Cephalosporin nitric oxide-donor prodrug Drug delivery P. aeruginosa A crystal violet staining technique and
CLSM
This compound leads to the near-complete
eradication of the biofilm community.
[70]
Polymeric derivative with glyco-polypep-
tide architecture
Drug delivery P. aeruginosa, S. aureus Cr ystal violet method, static chamber
system
This compound leads to the eradication of
established clinically relevant biofilms.
[71]
Clomiphene citrate and Auranofin5FDA-approved drugs P. aeruginosa Formation of biofilms on glass discs and
CLSM
The combination therapy showed antibi-
ofilm activity
[72]
Exopolysaccharide biosynthetic glycoside
hydrolases
Glycoside hydrolases P. aeruginosa CLSM, microtiter dish biofilm assay The addition of enzyme to colistin
decreased bacterial count.
[73]
HBED Iron chelators P. aeruginosa Microtiter plate and flow cell biofilm assay Combination therapy significantly increased
the effect of colistin microcolony killing
and leads to the almost complete removal
of the biofilm.
[74]
EDTA Iron chelators Colistin-resistant K. pneumonia Crystal violet assay and catheter-related
biofilm infection mouse model
Combination therapy remarkably reversed
colistin resistance in both planktonic and
mature biofilms of colistin-resistant and
eradicated colistin-resistant bacteria from
catheter-related biofilm infections.
[75]
CHIR-090 6LpxC inhibitors P. aeruginosa Bead biofilm assay, biofilms in flow cell
chambers, and mouse biofilm implant
model of infection
The combination therapy, at sub-inhibitory
concentrations, indicated synergistic
activity and suppressed the formation of
biofilm of colistin-tolerant bacteria.
[76]
Maipomycin 1Natural compound A. baumannii MTT assay and CLSM analysis Maipomycin increased the antibiofilm
activity of colistin.
[77]
Nutrient dispersion compounds Nutrient dispersion P. aeruginosa CLSM The combination therapy leads to the
remarkably significant decrease in the
live bacterial population.
[78]
PFK-158 4PFKFB3 inhibitor Colistin-resistant GNB Biofilm formation assay and SEM The combination therapy significantly
reduced biofilm formation and decreased
the cell arrangement density of biofilm.
[79]
Furanone C-30 3QS inhibitors Colistin-resistant GNB Biofilm formation inhibition assays and
standardized invitro biofilm mode
The combination therapy suppressed
the formation of bacterial biofilm and
showed a better eradication effect on
established biofilm than monotherapy.
[80]
N-(2-pyrimidyl) butanamide 8QS inhibitors P. aeruginosa CLSM, 24 well microtiter plates containing
BBM and AHL analogs
Synergistic antibiofilm activity was
detected under both anaerobic and aero-
bic conditions.
[81]
Ultrasound patches Ultrasound P. aeruginosa Filter-biofilms Significantly enhanced the bacterial killing
of the biofilm community.
[82]
Brazilian Journal of Microbiology
1 3
Aguilera-Correa etal. used bone-targeted mesoporous
silica nanoparticles that were functionalized with gelatin/
colistin coating. The nanoparticles significantly reduced
the number of MRSA in the bone just 24h after only one
dose. It is noteworthy to mention that S. aureus by secre-
tion of different enzymes, such as cysteine proteases, ser-
ine proteases, and metalloproteases, could degrade gelatin
coating and accelerate the delivery of antibiotics from drug
platforms on the infected bone [98]. In another study, the
authors used mesoporous silica nanoparticles that have been
loaded with moxifloxacin and further functionalized with
colistin and Arabic gum. This nanosystem showed high
affinity toward the biofilm community of E. coli because
of Arabic gum coating and antibacterial activity because of
the colistin disaggregating effect and moxifloxacin bacteri-
cidal effect. The nanosystem, in a short time, could release
large amounts of moxifloxacin; therefore, it could lead to
the preparation of a high concentration of antibiotics nearby
bacteria and in the site of infection from a low quantity of
nanoparticles that might decrease the potential side effects
associated with other administration routes. Additionally,
colistin can directly kill bacteria because this antibiotic is
easily absorbed on the surface of nanoparticles. Interest-
ingly, Arabic gum improves the adsorption capacity of nano-
particles, potentially diminishing the final nanoparticle dose
that would be required during the treatment if colistin alone
was to be used [96].
Finally, in a recently published study, microcontainers,
reservoir-based microdevices, were co-loaded with colistin
and ciprofloxacin for inhibition of P. aeruginosa biofilm.
The results of this study showed that co-loaded microcon-
tainers are superior to monotherapy and completely killed all
of the bacteria in the planktonic community. Furthermore,
antibiotics in microcontainers work significantly faster (just
five hours) than simple perfusion of antibiotics in biofilm.
The authors proposed that this effect is caused by the burst
release of the antibiotics from the microcontainers, which
creates an immediate high concentration of antibiotics at the
local site of infection and ultimately more destroyed biomass
[99].
Therefore, as mentioned in this section, different drug
platforms, such as liposomes, PLGA, and microcontainers,
have an acceptable capacity as delivery systems for inhibi-
tion of bacterial biofilm by reaching immediate high local
colistin concentrations at the site of infection, especially in
patients with CF and osteomyelitis. Additionally, the use
of drug platforms decreases the desired dose of colistin,
therefore reducing the side effects of this antibiotic. Drug
platforms could be functionalized by different antibacte-
rial agents such as natural compounds, mucin-degrading
enzymes, metal nanoparticles, and antibiotics; thus, the use
of drug platforms can enhance the antibiofilm activity of
colistin. Altogether, drug platforms containing colistin may
Table 1 (continued)
Antibacterial agents Class Bacterium Methods Outcome and comments Reference
Low-frequency ultrasound Ultrasound Pan-resistant biofilms of A. baumannii 24 well microtiter plates Colistin/vancomycin low-frequency ultra-
sound combination therapy decreased the
count of bacteria in biofilms after 8 h and
a continuing decline until 24 h.
[83]
QS, quorum-sensing inhibitor; CR, carbapenem-resistant; MBECs, minimal biofilm eradication concentrations; CLSM, confocal laser-scanning microscopy; DD, disc diffusion; GNB, Gram-neg-
ative bacilli; XDR, extensively drug-resistant; MTT, methythiazolyl tetrazolium assay; HBED, N, N'-bis (2-hydroxybenzyl) ethylenediamine-N, N'-diacetic acid (iron chelators); SEM, scanning
electron microscopy; BBM, basic medium for biofilm formation; AHL, N-acyl homoserine lactone
1 This compound was isolated from rare actinomycetes strain Kibdelosporangium phytohabitans XY-R10
2 The authors reported that the DD method and broth growth-based assays may not be good predictors of antibiotic susceptibility in biofilms
3 QS inhibitors
4 6-phosphofructo2-kinase/fructose-2, 6-bisphosphatase 3 (PFKFB3) inhibitor
5 Compounds with antibacterial and antibiofilm activities that are commercially available
6 LpxC inhibitors
7 Alginate oligomer
8 QS inhibitors
Brazilian Journal of Microbiology
1 3
be an interesting strategy for the inhibition and removal of
bacterial biofilms; however, further studies are required
to identify the precise mechanism underlying the efficient
removal of biofilms by these platforms.
In the end, other drug platforms, such as colistin-loaded
human albumin nanoparticles, bi-functional alginate oligo-
saccharide–polymixin conjugates, and colistin-conditioned
surfaces, showed promising results for inhibition of bacterial
biofilms (Table1) [3, 57, 100].
N‑Acetylcysteine
N-Acetylcysteine (NAC) is commonly administered as an
antioxidant due to the ability of the free thiol group to react
with nitrogen species and reactive oxygen by constituting
a precursor of intracellular glutathione. Additionally, NAC
is known as a mucolytic agent; therefore, this compound
clears thick mucus from the lungs and is used in combination
therapy with antibiotics for the treatment of lower respira-
tory tract infections [101, 102]. The results of the recently
published studies also showed that NAC could destroy the
matrix architecture of bacterial biofilm and enhance biofilm
breakdown. As mentioned, NAC is a nebulized mucolytic
agent and is used for the management of patients with CF.
These patients are mostly infected with MDR Gram-negative
bacteria such as P. aeruginosa and Stenotrophomonas malt-
ophilia, and the biofilm community of these bacteria is a
main problem in CF patients. Altogether, colistin is used as
a last resort in these patients, and combination therapy with
this antibiotic and NAC for removing MDR bacterial biofilm
could be an applicable therapeutic approach (Fig.1).
In this regard, Aksoy etal. reported that NAC reduced
the minimum biofilm inhibitory concentration (MBIC) of
different antibiotics such as colistin against E. coli, Proteus
mirabilis, and Pseudomonas putida [103]. In another experi-
ment, 18 S. maltophilia were collected, and the effect of the
colistin-NAC combination therapy was evaluated against
these bacteria. The combination therapy indicated syner-
gism against the colistin-resistant strains, suggesting that
NAC could antagonize the mechanisms involved in colistin
resistance. Additionally, a dose-dependent potentiation of
colistin activity at sub-MIC concentrations by NAC was also
clearly observed against S. maltophilia biofilms [104]. In
line with these results, Polline and colleagues reported a sig-
nificant synergistic antibiofilm function of N-acetylcysteine
(8000mg/L) plus colistin (8mg/L) against colistin-suscep-
tible and colistin-resistant strains [101].
Noteworthy, an artificial sputum medium (ASM) model
was used in two studies in order to mimic the bacterial bio-
film environmental conditions experienced in CF mucus. In
one of these studies, monotherapy with NAC (8000mg/L)
indicated strain-dependent and limited antibiofilm activity
against P. aeruginosa. However, the use of colistin (2 to
32mg/L) plus NAC (8000mg/L) demonstrated a relevant
antibiofilm synergism against all strains. Furthermore, this
combination therapy also demonstrated a clear synergism
against bacterial biofilms grown in ASM. Noteworthy, the
colistin concentration that allowed observation of synergism
was much higher (i.e., 32 × the MIC) in the ASM model in
comparison to the Nunc-TSP lid system. In this regard, the
authors proposed that the requirement for a higher dosage of
colistin could be due to the strong ionic interactions of this
antibiotic with ASM components such as mucin and extra-
cellular DNA. In addition, the results showed that treatment
of bacterial planktonic cultures with NAC could reduce the
virulence of P. aeruginosa, such as anaerobic respiration,
zinc starvation response, and flagellum-mediated motility.
Hence, pretreatment with NAC could prevent biofilm forma-
tion and lung infection [105].
In line with these results, Aiyer etal. evaluated the
impact of synergistic and additive colistin-NAC combi-
nation therapy in an ASM model using lung macrophages
and bronchial cells to model Achromobacter xylosoxidans
infection. Noteworthy, this bacterium is a Gram-negative
bacillus that has been related to chronic colonization in
CF and antibiotic resistance. The findings of this study
showed that combination therapy is well tolerated by both
cells and could lead to the synergistic and remarkable
decrease in bacterial counts [106]. These data indicated
that the antibiofilm synergism of colistin-NAC combina-
tions against P. aeruginosa strains is also preserved under
environmental conditions mimicking the CF mucus, which
is promising for clinical applications.
Noteworthy, NAC itself can suppress bacterial pro-
liferation and growth by preventing cysteine utilization
by bacterial cells and reducing the formation of EPS and
polysaccharides in many bacterial species [107]. NAC can
destabilize the biofilm structure by interacting with the
main components in the biofilm matrix or by chelating
magnesium and calcium [108]. As mentioned in previous
parts, NAC, at concentrations achievable by topical admin-
istration, could suppress the biofilm community of CF-
associated Gram-negative bacteria and revert the colistin-
resistant phenotype. In this setting, NAC might revert the
colistin resistance phenotype and remarkably improve the
efficacy of this antibiotic against bacterial biofilm. Nev-
ertheless, the report of exact interactions between colistin
and NAC that lead to synergism in combination therapy
is not easy to hypothesize due to the relevant knowledge
gaps on the mechanisms of action of both compounds.
In this regard, further studies with a focus on NAC and
colistin interactions are encouraged because understanding
the mechanisms of such a synergism would be relevant to
the discovery of new antibacterial agents for inhibition of
Brazilian Journal of Microbiology
1 3
MDR bacterial biofilm in different infections such as CF
and catheter-associated infections.
Natural compounds
Recently published studies have reported that different nat-
ural products or natural compounds (NCs) not only could
reduce the formation of biofilm but also could eradicate
an established biofilm structure [109111]. Furthermore,
probably a combination treatment of NCs with conven-
tional antibiotics, because of their increasing effective-
ness and potency while minimizing the toxicity and dosage
of antibiotics and reducing the likelihood of developing
resistant strains, should be considered as an applicable
therapeutic approach to manage MDR and biofilm-associ-
ated infection [112]. In this regard, the results of recently
published studies indicated that different NCs such as
resveratrol, chrysin, kaempferol, plumbagin, naringenin,
cinnamaldehyde, thymol, and capsaicin could enhance the
efficacy of colistin against biofilms of Gram-negative bac-
teria, even colistin-resistant strains [113120].
Noteworthy, inhibition of QS in Gram-negative bacte-
ria, especially P. aeruginosa, was considered by research-
ers as a promising therapeutic approach for inhibition
of bacterial biofilm and infections [121]. In this regard,
different NCs, such as cinnamaldehyde, could suppress
the expression of Las-, Rhl-, and PQS in P. aeruginosa.
The P. aeruginosa QS system is comprised of three hier-
archically integrated QS systems, Las, Rhl, and PQS, to
manage the expression of different virulence factors and
biofilm-related genes of this bacterium [118, 119, 122].
Therefore, NCs, by suppressing the QS system in bacteria,
could inhibit the formation of bacterial biofilm and make
them more susceptible to colistin.
In addition to QS inhibition, NCs could reduce the bio-
film formation by inhibition of bacterial attachment in the
first step of the switch of planktonic bacteria to a biofilm
phenotype. In this manner, the results of a study showed
that chrysin, a component of honey, could downregulate
the expression of csuA/B and katE [120]. Noteworthy, the
chaperoneusher pili (Csu) assembly system, including
transport proteins (CsuC and CsuD) and pilin subunits
(CsuA/B, CsuA, CsuB, and CsuE), is a main player in A.
baumannii adhesion to the medical appliances surfaces
[120, 123]. Moreover, RpoS has an association with the
formation of established biofilm by inducing motility-
related genes and by suppressing EPS synthesis, and katE
is a reporter gene of RpoS [120, 124, 125]. To this end,
when the expression of the mentioned genes is downregu-
lated by colistin-chrysin combination therapy, the forma-
tion of biofilm is also inhibited.
Furthermore, NCs showed interaction with the outer
membrane and altered the potential of the cell membrane.
Fig. 1 The use of combination therapy for enhancement of colistin activity against bacterial biofilm
Brazilian Journal of Microbiology
1 3
For example, the findings of a study showed that thymol
can enhance the permeability of membranes to overcome
colistin resistance. Capsaicin can also destroy the perme-
ability of the outer membrane, which could enable hydro-
phobic capsaicin molecules to pass through the LPS in the
outer layer of bacteria and reach their target, thus playing
a synergistic role [113, 114]. Therefore, NCs are permea-
bilizers and should be considered a promising adjuvant to
colistin against Gram-negative bacteria. Finally, it should
be noted that NCs could suppress the expression of efflux
pumps, and this possible mechanism should be evaluated in
future studies as a possible synergistic effect in the colistin-
NCs combination therapy [77]. Hence, NCs can improve
the efficacy of colistin against bacterial biofilm and over-
come colistin resistance; therefore, the synergy between
colistin and NCs warrants further experimental evaluation
and confirmation using different animal models of infection
and different dosage combinations. However, some disad-
vantages, such as poor stability and solubility, hinder the
use of NCs in clinical settings. To this end, the use of drug
delivery platforms can overcome these physicochemical
barriers [119]. Additionally, some of the NCs are used as
antioxidant agents, while the production of reactive oxygen
species (ROS) is one of the important antibacterial mecha-
nisms mediated by colistin [126]. Therefore, co-administra-
tion of NCs that remove ROS could increase the persister
cells, and this issue should be evaluated in further studies.
Antimicrobial peptides
Antimicrobial peptides (AMPs), host defense peptides that
mainly consist of protein fragments, are produced by both
prokaryotes and eukaryotes and act as a first line of defense
for the immune system [127]. AMPs are broad-spectrum
antibacterial agents that kill bacteria rapidly with low cyto-
toxicity for eukaryotic cells [127, 128]. Additionally, these
agents have an antibiofilm capacity using different mecha-
nisms, such as disruption of membrane and EPS, downreg-
ulation of biofilm-associated genes, interference with cell
signaling, and inhibition of stringent response [129].
In this regard, researchers considered colistin-AMP com-
bination therapy a promising antibiofilm strategy. Morroni
etal. reported synergic activity for colistin-LL37 combina-
tion therapy against ESBL- and carbapenemase-producing and
mcr-1 carrying E. coli. Noteworthy, AMP LL-37 is a 37-amino
acid peptide that is proteolytically released from the human
cathelicidin hCAP-18. AMP LL-37 significantly reduced the
E. coli biofilm at 1 × MIC concentration, and the expression
of mcr1, a colistin-resistant-associated gene, did not affect the
function of this AMP [130]. The results of another study also
showed that a combination of colistin and different AMPs
such as LL-37 could decrease minimum biofilm eradication
concentration (MBEC) by eightfold [131]. Other authors also
reported that a combination of the sub-MIC concentration of
CRAMP (a cathelicidin-associated AMP) with colistin indi-
cated interesting synergistic activity against the P. aeruginosa
biofilm community and reduced the expression of QS-regu-
lated genes, resulting in inhibitory effects on QS-regulated
virulence phenotypes (pyocyanin and rhamnolipid) [132].
Furthermore, the findings of a recently published study dem-
onstrated that colistin, in combination with AMPs, including
citropin 1.1, MP temporin A, and tachyplesin I linear analogue,
have an additive or synergic function in the treatment of pre-
established S. aureus and P. aeruginosa single- and double-
species biofilm [133]. Finally, the combination of colistin with
melittin, an alpha-helical hydrophobic AMP, completely inhib-
ited the biofilm formation of MDR-strong biofilm producer A.
baumannii. Furthermore, the results of this study showed that
melittin decreased the expression of the bap gene. This phe-
nomenon decreases the accumulation of extracellular matrix
in the periphery of bacteria, which consequently enhances the
penetration of antibiotics into the cytoplasm. Besides, it is note-
worthy to mention that melittin can bind to the resistant gene
and inhibit its expression by its DNA binding activity [134].
It seems colistin-AMP combination therapy leads to syn-
ergistic effects due to the diversity of mechanisms of action
found in colistin and AMPs. For instance, colistin, after bind-
ing to the LPS of Gram-negative bacteria, leads to the disrup-
tion of the outer membrane, while AMPs interact with the
cytoplasmic membrane. Therefore, the combination of these
two antibacterial agents causes a perturbation of both the outer
and cytoplasmic membranes and could explain the synergistic
relation between colistin and different AMPs such as LL-37
[6, 130, 135]. Additionally, as mentioned, colistin-AMP com-
bination therapy could suppress the biofilm formation in P.
aeruginosa by downregulation of the rhl system gene, result-
ing in decreased expression of pyocyanin and rhamnolipid. It
should be noted that pyocyanin and rhamnolipid are important
virulence factors in P. aeruginosa that are mainly regulated by
the rhl system and manage the secretion of the extracellular
DNA in bacteria (eDNA) and play a vital role in the formation
and diffusion of biofilm [132, 136, 137]. Therefore, AMPs
could destroy the structure of biofilm by downregulating the
QS-associated gene and consequently improve the interactions
of colistin with biofilm. However, the studies in this field are
very limited, and further invivo studies are needed to evaluate
the synergistic activity of colistin and AMPs and determine the
possible clinical dose through pharmacokinetic experiments.
Phage
Bacteriophages (phages), viruses that infect bacteria, are
considered by scientists as an applicable approach for the
management of MDR bacteria. Additionally, recent studies
Brazilian Journal of Microbiology
1 3
have reported phages as antibacterial agents with inhibitory
effects against bacterial biofilm. However, some disadvan-
tages, such as the narrow host range and the development
of phage resistance, restrict the clinical usage of phages
[138]. To this end, the combination of phages and antibiot-
ics was considered by scientists to inhibit bacterial infection.
Colistin is one of these antibiotics that showed promising
inhibitory effects against MDR bacterial biofilm when used
in combination with phages and phage-derived enzymes.
Vashisth etal. evaluated the synergistic effects of Myophage
φAB182 in combination with different antibiotics such as
colistin against the biofilm community of MDR-A. bauman-
nii. The results of this study showed that φAB182 has the
highest synergy with colistin in comparison to other anti-
biotics such as ceftazidime, polymixin B, and cefotaxime.
Furthermore, colistin-phage combination therapy also eradi-
cated the biofilm of A. baumannii [139].
In line with these results, another investigation also
reported that colistin-phage T1245 combination therapy
not only could reduce bacterial density up to approximately
80% but also could reduce biomass and bacterial viability in
3-day established biofilms. After the combination therapy,
scanning microscopic evaluation showed visible alterations
in cell morphology, with membrane poration and cell lysis as
indicated by the presence of cell debris [140]. vWU2001 was
another phage that, in combination with colistin, remarkably
inhibited carbapenem-resistant A. baumannii in comparison
to monotherapy. This combination therapy also leads to a
significantly greater enhancement in G. mellonella survival
and in bacterial clearance, as compared with that of phage or
colistin alone [141]. Therefore, the use of colistin in combi-
nation with phages could be used for the inhibition of MDR-
A. baumannii biofilm. The exact interaction of phages and
colistin is not yet elucidated, but some possible mechanisms
have been reported by recent studies. For instance, colistin
interaction with the outer membrane of Gram-negative bac-
teria could increase both phage adsorption and phage DNA
injection [142]. Phage-antibiotic combination therapy may
lead to changes in bacterial morphology, rapid cell lysis, and
phage maturation [143]. Additionally, colistin could cause
cell clustering, and this phenomenon enhanced the phage’s
ability to travel on the adjoined cell surface, increasing
phage infection efficiency [144].
It is noteworthy to mention that phages produce the
endolysins during the final stages of the replication cycle to
cleave the bacterial cell wall and produce progeny virions.
Endolysins showed good inhibitory functions against Gram-
positive bacteria; however, the presence of the outer mem-
brane restricted their function against Gram-negative bacte-
ria [145, 146]. In this regard, colistin could be employed to
assist the endolysins in overcoming the impenetrability of
the outer membrane [146]. Finally, depolymerases that are
encoded by phages are responsible for destroying EPS, LPS,
and capsular polysaccharides (CPS) of the host bacteria dur-
ing phage invasion. Therefore, phages can destroy biofilm
structure and improve the penetration of the antibiotic to
the deeper layers of biofilm by inducing the synthesis of
enzymes such as polysaccharide depolymerase [147]. Hen
etal. used depolymerase Dpo71, derived from a baumannii
phage, in combination with colistin, for inhibition of the
biofilm community of MDR-A. baumannii. Dpo71 improved
the inhibitory effect of host immune cell activity against
bacteria and also acts as an adjuvant to assist or improve
the function of colistin. An exact evaluation indicated that
the enhanced bactericidal effect of colistin is attributed to
the improved outer membrane destabilization capacity and
binding rate to bacteria after stripping off the bacterial cap-
sule by Dpo71. Moreover, the combination of Dpo71 could
remarkably improve the colistin activity against biofilm and
enhance the survival rate of A. baumannii-infected Galleria
mellonella. The results of the microscopy evaluation showed
that Dpo71 could significantly destroy the biofilms but was
not able to decrease the count of viable cells in the biofilms.
Colistin-Dpo71 combination therapy significantly reduced
the count of viable bacterial cells and residual biomass of
biofilm compared with monotherapy. It seems that the antib-
iofilm activity of his combination therapy is associated with
the improved colistin penetration within the biofilm matrix
after the EPS depolymerization by the Dpo71 [148].
Therefore, the combined use of colistin and phage depoly-
merase should be considered for the inhibition of the biofilm
of MDR bacteria. However, knowledge of the exact interac-
tion of extracellular structures of Gram-negative bacteria
cleavage by phage depolymerases is still largely missing.
Additionally, the biofilm’s susceptibility to phage depoly-
merase treatments varied, depending on the bacterial strains,
the activity of the depolymerase enzyme, and the type of
bacteria. Therefore, although colistin-phage combination
therapy may represent promising treatment strategies for
managing MDR bacteria, more confirmatory studies in this
field are required.
Conclusion
As mentioned in the previous sections, the use of combina-
tion therapy could boost the inhibitory effects of colistin
against the MDR bacterial biofilm community. To this end,
the use of different antibacterial agents, antibiotics, and drug
platforms in combination with colistin could suppress bio-
film formation and remove established biofilm. Additionally,
combination therapy reduces the chance of acquiring colistin
resistance and decreases the required dose of this antibiotic
at the site of infection. Therefore, colistin-based combination
therapy could open new frontiers in the treatment of biofilm-
related infections; however, the exact mechanism underlying
Brazilian Journal of Microbiology
1 3
the efficient removal of biofilms by combination therapy has
not yet been reported. To this end, further investigation of
preclinical and clinical studies as well as bio-toxicity evalu-
ation for the human body is needed before the clinical usage
of colistin-based combination therapy for the management
of biofilm-associated infections.
Abbreviations MDR:Multi-drug resistant; LPS:Lipopolysaccharides;
MIC:Minimum inhibitory concentration; ESBL:Extended-spectrum-
β-lactamase; XDR:Extensively drug-resistant; QS:Quorum sensing;
MRSA:Methicillin-resistant Staphylococcus aureus; NAC:N-acetyl-
cysteine; CF:Cystic fibrosis; MBIC:Minimum biofilm inhibitory con-
centration; ASM:Artificial sputum medium; NCs:Natural compounds;
MBEC:Minimum biofilm eradication concentration
Acknowledgements We greatly appreciate the input from the BioRender
team (BioRender.com) for their collaboration with us in the figure design.
Author contribution AJ conceived and designed the study. RA, MA
MS, MM, MA, and RL contributed to comprehensive research and
wrote the paper. Notably, all authors have read and approved the
manuscript.
Data availability The authors confirm that the data supporting the find-
ings of this study is available within the article.
Declarations
Ethics approval and consent to participate Not applicable.
Consent for publication Not applicable.
Conflict of interest The authors declare no competing interests.
References
1. Sharma J, Sharma D, Singh A, Sunita K (2022) Colistin resist-
ance and management of drug resistant infections. Can J Infect
Dis Med Microbiol 2022:1–10
2. Gurjar M (2015) Colistin for lung infection: an update. J Inten-
sive Care 3:3
3. Scutera S, Argenziano M, Sparti R, Bessone F, Bianco G, Bas-
tiancich C etal (2021) Enhanced antimicrobial and antibiofilm
effect of new colistin-loaded human albumin nanoparticles. Anti-
biotics (Basel, Switzerland) 10(1):57
4. Ayoub Moubareck C (2020) Polymyxins and bacterial mem-
branes: a review of antibacterial activity and mechanisms of
resistance. Membranes 10(8):181
5. Bergen PJ, Bulman ZP, Landersdorfer CB, Smith N, Lenhard
JR, Bulitta JB etal (2015) Optimizing polymyxin combina-
tions against resistant Gram-negative bacteria. Infect Dis Ther
4:391–415
6. Bialvaei AZ, Samadi KH (2015) Colistin, mechanisms and preva-
lence of resistance. Curr Med Res Opin 31:707–721
7. Lora-Tamayo J, Murillo O, Ariza J (2019) Clinical use of
colistin in biofilm-associated infections. Adv Exp Med Biol
1145:181–195
8. Høiby N, Bjarnsholt T, Givskov M, Molin S, Ciofu O (2010)
Antibiotic resistance of bacterial biofilms. Int J Antimicrob
Agents 35:322–332
9. Park NH, Lee SJ, Lee EB, Birhanu BT, Park SC (2021) Colis-
tin induces resistance through biofilm formation, via increased
phoQ expression, in avian pathogenic Escherichia coli. Patho-
gens (Basel, Switzerland) 10(11):1525
10. Bergen PJ, Landersdorfer CB, Zhang J, Zhao M, Lee HJ, Nation
RL etal (2012) Pharmacokinetics and pharmacodynamics of
‘old’ polymyxins: what is new? Diagn Microbiol Infect Dis
74:213–223
11 Falagas ME, Rafailidis PI, Kasiakou SK, Hatzopoulou P, Mich-
alopoulos A (2006) Effectiveness and nephrotoxicity of colistin
monotherapy vs. colistin-meropenem combination therapy for
multidrug-resistant Gram-negative bacterial infections. Clin
Microbiol Infect: Off Publ Eur Soc Clin Microbiol Infect Dis
12:1227–30
12. Batirel A, Balkan II, Karabay O, Agalar C, Akalin S, Alici O etal
(2014) Comparison of colistin-carbapenem, colistin-sulbactam,
and colistin plus other antibacterial agents for the treatment of
extremely drug-resistant Acinetobacter baumannii bloodstream
infections. Eur J Clin Microbiol Infect Dis: Off Publ Eur Soc Clin
Microbiol 33:1311–1322
13. Shariati A, Azimi T, Ardebili A, Chirani A, Bahramian A, Por-
mohammad A etal (2018) Insertional inactivation of oprD in
carbapenem-resistant Pseudomonas aeruginosa strains isolated
from burn patients in Tehran, Iran. New Microbes New Infect
21:75–80
14. Sharma U, Vipra A, Channabasappa S (2018) Phage-derived
lysins as potential agents for eradicating biofilms and persisters.
Drug Discov Today 23:848–856
15. Obaid RF, Kadhim Hindi NK, Kadhum SA, Jafaar Alwaeli
LA, Jalil AT (2022) Antibacterial activity, anti-adherence and
anti-biofilm activities of plants extracts against Aggregatibacter
actinomycetemcomitans: an invitro study in Hilla City, Iraq.
Caspian J Environ Sci 20:367–372
16. Sharahi JY, Azimi T, Shariati A, Safari H, Tehrani MK, Hashemi
A (2019) Advanced strategies for combating bacterial biofilms.
J Cell Physiol 234:14689–14708
17. Donlan RM (2001) Biofilm formation: a clinically relevant
microbiological process. Clin Infect Dis 33:1387–1392
18. Sadekuzzaman M, Yang S, Mizan M, Ha S (2015) Current and
recent advanced strategies for combating biofilms. Comp Rev
Food Sci Food Safe 14:491–509
19. Elbarasi A (2014) Identification and characterization of msaB
gene involved in biofilm formation and virulence in Staphylococ-
cus aureus
20. Alhamadani Y, Oudah A (2022) Study of the Bacterial Sensitivity
to different Antibiotics which are isolated from patients with UTI
using Kirby-Bauer Method. J Biomed Biochem 1(2):1–6.https://
doi. org/ 10. 57238/ jbb. 2022. 19387
21. Donlan RM (2000) Role of biofilms in antimicrobial resistance.
ASAIO J 46:S47–S52
22. AlChalabi R, Al-Rahim A, Omer D, Suleiman AA (2022) Immu-
noinformatics design of multi-epitope peptide-based vaccine
against Haemophilus influenzae strain using cell division protein.
Netw Model Anal Health Inform Bioinforma 12(1):1
23. Howell-Jones R, Wilson M, Hill KE, Howard A, Price PE,
Thomas DW (2005) A review of the microbiology, antibiotic
usage and resistance in chronic skin wounds. J Antimicrob
Chemother 55:143–149
24. Hoyle BD, Wong CK, Costerton JW (1992) Disparate efficacy of
tobramycin on Ca2+-, Mg2+-, and HEPES-treated Pseudomonas
aeruginosa biofilms. Can J Microbiol 38:1214–1218
25. Duguid IG, Evans E, Brown MR, Gilbert P (1992) Effect of bio-
film culture upon the susceptibility of Staphylococcus epider-
midis to tobramycin. J Antimicrob Chemother 30:803–810
26. Christensen BB, Sternberg C, Andersen JB, Eberl L, Møller
S, Givskov M etal (1998) Establishment of new genetic traits
Brazilian Journal of Microbiology
1 3
in a microbial biofilm community. Appl Environ Microbiol
64:2247–2255
27. Elias S, Banin E (2012) Multi-species biofilms: living with
friendly neighbors. FEMS Microbiol Rev 36:990–1004
28. Fux C, Costerton J, Stewart P, Stoodley P (2005) Th1 and Th2-
weighted immune responses. Trends Microbiol 1:34–40
29. Guerra MES, Destro G, Vieira B, Lima AS, Ferraz LFC, Hakans-
son AP etal (2022) Klebsiella pneumoniae biofilms and their role
in disease pathogenesis. Front Cell Infect Microbiol 12:877995
30. Moghadam MT, Shariati A, Mirkalantari S, Karmostaji A (2020)
The complex genetic region conferring transferable antibiotic
resistance in multidrug-resistant and extremely drug-resistant
Klebsiella pneumoniae clinical isolates. New Microbes New
Infect 36:100693
31. Neut D, Tijdens-Creusen EJ, Bulstra SK, van der Mei HC,
Busscher HJ (2011) Biofilms in chronic diabetic foot ulcers—a
study of 2 cases. Acta Orthop 82:383–385
32. Zhao G, Usui ML, Lippman SI, James GA, Stewart PS, Fleckman
P etal (2013) Biofilms and inflammation in chronic wounds. Adv
Wound Care 2:389–399
33. Kathju S, Nistico L, Hall-Stoodley L, Post JC, Ehrlich GD,
Stoodley P (2009) Chronic surgical site infection due to suture-
associated polymicrobial biofilm. Surg Infect 10:457–461
34. Copur B, Dosler S, Aktas Z, Basaran S, Simsek-Yavuz S, Caga-
tay A etal (2022) Invitro activities of antibiotic combinations
against mature biofilms of ventilator-associated pneumonia iso-
lates. Future Microbiol 17:1027–1042
35. Taşkın Kafa AH, Hasbek M (2022) Synergistic efficacy of mero-
penem, ciprofloxacin and colistin antibiotics against planktonic
and biofilm forms of Myroides odoratimimus bacterial isolates.
Indian J Med Microbiol 40:399–403
36. Memar MY, Adibkia K, Farajnia S, Samadi Kafil H, Khalili Y,
Azargun R etal (2021) In-vitro effect of imipenem, fosfomycin,
colistin, and gentamicin combination against carbapenem-resist-
ant and biofilm-forming Pseudomonas aeruginosa isolated from
burn patients. Iran J Pharm Res: IJPR 20:286–296
37. Geladari A, Simitsopoulou M, Antachopoulos C, Roilides E
(2019) Dose-dependent synergistic interactions of colistin with
rifampin, meropenem, and tigecycline against carbapenem-
resistant Klebsiella pneumoniae biofilms. Antimicrob Agents
Chemother 63(3):10–1128
38. Lora-Tamayo J, Murillo O, Bergen PJ, Nation RL, Poudyal A,
Luo X etal (2014) Activity of colistin combined with doripenem
at clinically relevant concentrations against multidrug-resistant
Pseudomonas aeruginosa in an invitro dynamic biofilm model.
J Antimicrob Chemother 69:2434–2442
39. Ribera A, Benavent E, El-Haj C, Gomez-Junyent J, Tubau F,
Rigo-Bonnin R, Murillo O (2019) Comparative antibiofilm effi-
cacy of meropenem alone and in combination with colistin in
an invitro pharmacodynamic model by extended-spectrum-β-
lactamase-producing Klebsiella pneumoniae. Antimicrob Agents
Chemother 63(11): 10–1128
40. Gómez-Junyent J, Murillo O, Yu HH, Azad MAK, Wickremas-
inghe H, Rigo-Bonnin R etal (2021) Invitro pharmacokinetics/
pharmacodynamics of continuous ceftazidime infusion alone and
in combination with colistin against Pseudomonas aeruginosa
biofilm. Int J Antimicrob Agents 57:106246
41. Gómez-Junyent J, Benavent E, Sierra Y, El Haj C, Soldevila L,
Torrejón B etal (2019) Efficacy of ceftolozane/tazobactam, alone
and in combination with colistin, against multidrug-resistant
Pseudomonas aeruginosa in an invitro biofilm pharmacody-
namic model. Int J Antimicrob Agents 53:612–619
42. Pamp SJ, Gjermansen M, Johansen HK, Tolker-Nielsen T (2008)
Tolerance to the antimicrobial peptide colistin in Pseudomonas
aeruginosa biofilms is linked to metabolically active cells, and
depends on the pmr and mexAB-oprM genes. Mol Microbiol
68:223–240
43. Pletzer D, Hancock RE (2016) Antibiofilm peptides: potential as
broad-spectrum agents. J Bacteriol 198:2572–2578
44 Kolpen M, Appeldorff CF, Brandt S, Mousavi N, Kragh KN,
Aydogan S etal (2016) Increased bactericidal activity of colistin
on Pseudomonas aeruginosa biofilms in anaerobic conditions.
Pathog Dis 74:ftv086
45. Haagensen J, Verotta D, Huang L, Engel J, Spormann AM, Yang
K (2017) Spatiotemporal pharmacodynamics of meropenem- and
tobramycin-treated Pseudomonas aeruginosa biofilms. J Antimi-
crob Chemother 72:3357–3365
46. Mahamad Maifiah MH, Zhu Y, Tsuji BT, Creek DJ, Velkov T,
Li J (2022) Integrated metabolomic and transcriptomic analyses
of the synergistic effect of polymyxin-rifampicin combination
against Pseudomonas aeruginosa. J Biomed Sci 29:89
47. Song JY, Cheong HJ, Noh JY, Kim WJ (2015) Invitro compari-
son of anti-biofilm effects against carbapenem-resistant acine-
tobacter baumannii: imipenem, colistin, tigecycline, rifampicin
and combinations. Infect Chemother 47:27–32
48. Bergen PJ, Forrest A, Bulitta JB, Tsuji BT, Sidjabat HE, Pater-
son DL etal (2011) Clinically relevant plasma concentrations
of colistin in combination with imipenem enhance pharma-
codynamic activity against multidrug-resistant Pseudomonas
aeruginosa at multiple inocula. Antimicrob Agents Chemother
55:5134–5142
49. Forrest GN, Tamura K (2010) Rifampin combination therapy for
nonmycobacterial infections. Clin Microbiol Rev 23:14–34
50. Mataraci Kara E, Ozbek CB (2018) Investigation of the effects
of various antibiotics against Klebsiella pneumoniae biofilms on
invitro catheter model. J Chemother (Florence, Italy) 30:82–88
51. Sato Y, Ubagai T, Tansho-Nagakawa S, Yoshino Y, Ono Y (2021)
Effects of colistin and tigecycline on multidrug-resistant Acine-
tobacter baumannii biofilms: advantages and disadvantages of
their combination. Sci Rep 11:11700
52 Ayoub Moubareck C, Hammoudi Halat D (2020) Insights into
Acinetobacter baumannii: a review of microbiological, virulence,
and resistance traits in a threatening nosocomial pathogen. Anti-
biotics (Basel, Switzerland) 9(3):119
53. Lin MF, Lin YY, Lan CY (2017) Contribution of EmrAB efflux
pumps to colistin resistance in Acinetobacter baumannii. J
Microbiol (Seoul, Korea) 55:130–136
54. Cheng J, Kesavan DK, Vasudevan A, Cai W, Wang H, Su Z etal
(2020) Genome and transcriptome analysis of A. baumannii’s
“transient” increase in drug resistance under tigecycline pressure.
J Glob Antimicrob Resist 22:219–25
55. Hengzhuang W, Song Z, Ciofu O, Onsøyen E, Rye PD, Høiby
N (2016) OligoG CF-5/20 Disruption of mucoid Pseudomonas
aeruginosa biofilm in a murine lung infection model. Antimicrob
Agents Chemother 60:2620–2626
56. Pritchard MF, Powell LC, Jack AA, Powell K, Beck K, Florance
H, Thomas DW (2017) A low-molecular-weight alginate oligo-
saccharide disrupts pseudomonal microcolony formation and
enhances antibiotic effectiveness. Antimicrob Agents chemother
61(9): 10–1128
57. Stokniene J, Powell LC, Aarstad OA, Aachmann FL, Rye PD,
Hill KE etal (2020) Bi-functional alginate oligosaccharide-pol-
ymyxin conjugates for improved treatment of multidrug-resistant
gram-negative bacterial infections. Pharmaceutics 12(11):1080
58. Sanchez CJ Jr, Akers KS, Romano DR, Woodbury RL, Hardy
SK, Murray CK etal (2014) D-amino acids enhance the activity
of antimicrobials against biofilms of clinical wound isolates of
Staphylococcus aureus and Pseudomonas aeruginosa. Antimi-
crob Agents Chemother 58:4353–4361
Brazilian Journal of Microbiology
1 3
59. Ozbek B, Mataraci E (2013) Invitro effectiveness of colistin,
tigecycline and levofloxacin alone and combined with clarithro-
mycin and/or heparin as lock solutions against embedded Acine-
tobacter baumannii strains. J Antimicrob Chemother 68:827–830
60. Wang Y, Li C, Wang J, Bai N, Zhang H, Chi Y etal (2022)
The efficacy of colistin combined with amikacin or levofloxacin
against Pseudomonas aeruginosa biofilm infection. Microbiol
Spectr 10:e0146822
61. Moshynets OV, Baranovskyi TP, Cameron S, Iungin OS, Pok-
holenko I, Jerdan R etal (2022) Azithromycin possesses bio-
film-inhibitory activity and potentiates non-bactericidal colistin
methanesulfonate (CMS) and polymyxin B against Klebsiella
pneumonia. PLoS One 17:e0270983
62. Boncompagni SR, Micieli M, Di Maggio T, Aiezza N, Antonelli
A, Giani T etal (2022) Activity of fosfomycin/colistin combina-
tions against planktonic and biofilm Gram-negative pathogens.
J Antimicrob Chemother 77:2199–2208
63. Mei Q, Geng S, Fang X, He Y, Liu L, Xu M etal (2019) [In vitro
activity of ceftazidime-avibactam combined with colistin against
extensively drug-resistant Pseudomonas aeruginosa]. Zhonghua
wei zhong bing ji jiu yi xue 31:1212–8
64. Ozbek B, Mataraci-Kara E (2016) Comparative invitro effica-
cies of various antipseudomonal antibiotics based catheter lock
solutions on eradication of Pseudomonas aeruginosa biofilms. J
Chemother (Florence, Italy) 28:20–24
65. Herrmann G, Yang L, Wu H, Song Z, Wang H, Høiby N etal
(2010) Colistin-tobramycin combinations are superior to mono-
therapy concerning the killing of biofilm Pseudomonas aerugi-
nosa. J Infect Dis 202:1585–1592
66. Zhou QT, Sun SP, Chan JG, Wang P, Barraud N, Rice SA etal
(2015) Novel inhaled combination powder containing amorphous
colistin and crystalline rifapentine with enhanced antimicrobial
activities against planktonic cells and biofilm of Pseudomonas
aeruginosa for respiratory infections. Mol Pharm 12:2594–2603
67. Zhang X, Zhao Y, Feng L, Xu M, Ge Y, Wang L etal (2021)
Combined With mefloquine, resurrect colistin active in colistin-
resistant Pseudomonas aeruginosa invitro and invivo. Front
Microbiol 12:790220
68. Field D, Seisling N, Cotter PD, Ross RP, Hill C (2016) Syner-
gistic Nisin-polymyxin combinations for the control of Pseu-
domonas biofilm formation. Front Microbiol 7:1713
69. Al Atya AK, Abriouel H, Kempf I, Jouy E, Auclair E, Vachée A
etal (2016) Effects of colistin and bacteriocins combinations on
the invitro growth of Escherichia coli strains from swine origin.
Probiotics Antimicrob Proteins 8:183–190
70. Soren O, Rineh A, Silva DG, Cai Y, Howlin RP, Allan RN etal
(2020) Cephalosporin nitric oxide-donor prodrug DEA-C3D
disperses biofilms formed by clinical cystic fibrosis isolates of
Pseudomonas aeruginosa. J Antimicrob Chemother 75:117–125
71. Mauro N, Schillaci D, Varvarà P, Cusimano MG, Geraci DM,
Giuffrè M etal (2018) Branched high molecular weight glyco-
polypeptide with broad-spectrum antimicrobial activity for the
treatment of biofilm related infections. ACS Appl Mater Inter-
faces 10:318–331
72. Torres NS, Montelongo-Jauregui D, Abercrombie JJ, Srinivasan
A, Lopez-Ribot JL, Ramasubramanian AK etal (2018) Antimi-
crobial and antibiofilm activity of synergistic combinations of
a commercially available small compound library with colistin
against Pseudomonas aeruginosa. Front Microbiol 9:2541
73. Baker P, Hill PJ, Snarr BD, Alnabelseya N, Pestrak MJ, Lee MJ
etal (2016) Exopolysaccharide biosynthetic glycoside hydrolases
can be utilized to disrupt and prevent Pseudomonas aeruginosa
biofilms. Sci Adv 2:e1501632
74 Mettrick K, Hassan K, Lamont I, Reid D (2020) The iron-chela-
tor, N, N’-bis (2-hydroxybenzyl) ethylenediamine-N, N’-diacetic
acid is an effective colistin adjunct against clinical strains of
biofilm-dwelling Pseudomonas aeruginosa. Antibiotics (Basel,
Switzerland) 9(4):144
75. Shein AMS, Wannigama DL, Higgins PG, Hurst C, Abe S,
Hongsing P etal (2021) Novel colistin-EDTA combination for
successful eradication of colistin-resistant Klebsiella pneumoniae
catheter-related biofilm infections. Sci Rep 11:21676
76. Tan JH, Vidaillac C, Yam JKH, Chua SL, Givskov M, Yang L
(2017) Invitro and invivo efficacy of an LpxC inhibitor, CHIR-
090, alone or combined with colistin against Pseudomonas aer-
uginosa biofilm. Antimicrob Agents Chemother 61(7):10–1128
77. Zhang J, Liang X, Zhang S, Song Z, Wang C, Xu Y, Maipomycin
A (2020) a novel natural compound with promising anti-biofilm
activity against Gram-negative pathogenic bacteria. Front Micro-
biol 11:598024
78. Sommerfeld Ross S, Fiegel J (2012) Nutrient dispersion enhances
conventional antibiotic activity against Pseudomonas aeruginosa
biofilms. Int J Antimicrob Agents 40:177–181
79. Chen L, Yu K, Chen L, Zheng X, Huang N, Lin Y etal (2021)
Synergistic activity and biofilm formation effect of colistin com-
bined with PFK-158 against colistin-resistant Gram-negative
bacteria. Infect Drug Resist 14:2143–2154
80. Zhang Y, Lin Y, Zhang X, Chen L, Xu C, Liu S etal (2021)
Combining colistin with furanone C-30 rescues colistin resist-
ance of gram-negative bacteria invitro and invivo. Microbiol
Spectr 9:e0123121
81. Furiga A, Lajoie B, El Hage S, Baziard G, Roques C (2015)
Impairment of Pseudomonas aeruginosa biofilm resistance to
antibiotics by combining the drugs with a new quorum-sensing
inhibitor. Antimicrob Agents Chemother 60:1676–1686
82. Kvich L, Christensen MH, Pierchala MK, Astafiev K, Lou-Moe-
ller R, Bjarnsholt T (2022) The combination of low-frequency
ultrasound and antibiotics improves the killing of invitro Staphy-
lococcus aureus and Pseudomonas aeruginosa biofilms. Antibiot-
ics (Basel, Switzerland) 11(11):1494
83. Liu X, Yin H, Weng CX, Cai Y (2016) Low-frequency ultrasound
enhances antimicrobial activity of colistin-vancomycin combina-
tion against pan-resistant biofilm of Acinetobacter baumannii.
Ultrasound Med Biol 42:1968–1975
84. Khoshbayan A, Shariati A, Razavi S, Baseri Z, Ghodousi A, Dar-
ban-Sarokhalil D (2022) Mutation in mgrB is the major colistin
resistance mechanism in Klebsiella pneumoniae clinical isolates
in Tehran, Iran. Acta Microbiol Immunol Hung 69(1):61–67
85. Roomi AB, Widjaja G, Savitri D, Turki Jalil A, Fakri Mustafa
Y, Thangavelu L etal (2021) SnO2:Au/carbon quantum dots
nanocomposites: synthesis, characterization, and antibacterial
activity. J Nanostruct 11:514–523
86. Obaid Saleh R, Olegovich Bokov D, Fenjan MN, Kamal Abdel-
basset W, Altimari US, Turki Jalil A etal (2022) Application
of aluminum nitride nanotubes as a promising nanocarriers for
anticancer drug 5-aminosalicylic acid in drug delivery system. J
Mol Liq 352:118676
87. Honarvari B, Karimifard S, Akhtari N, Mehrarya M, Moghaddam
ZS, Ansari MJ etal (2022) Folate-targeted curcumin-loaded
niosomes for site-specific delivery in breast cancer treatment: in
silico and invitro study. Molecules (Basel, Switzerland) 27:4634
88. Sans-Serramitjana E, Jorba M, Pedraz JL, Vinuesa T, Viñas
M (2017) Determination of the spatiotemporal dependence of
Pseudomonas aeruginosa biofilm viability after treatment with
NLC-colistin. Int J Nanomed 12:4409–4413
89. Sans-Serramitjana E, Fusté E, Martínez-Garriga B, Merlos
A, Pastor M, Pedraz JL etal (2016) Killing effect of nanoen-
capsulated colistin sulfate on Pseudomonas aeruginosa from
cystic fibrosis patients. J Cyst Fibros: Off J Eur Cyst Fibros Soc
15:611–618
90. d’Angelo I, Casciaro B, Miro A, Quaglia F, Mangoni ML, Ungaro
F (2015) Overcoming barriers in Pseudomonas aeruginosa lung
Brazilian Journal of Microbiology
1 3
infections: engineered nanoparticles for local delivery of a cati-
onic antimicrobial peptide. Colloids Surf, B 135:717–725
91. Wang YY, Lai SK, So C, Schneider C, Cone R, Hanes J
(2011) Mucoadhesive nanoparticles may disrupt the protec-
tive human mucus barrier by altering its microstructure. PLoS
One 6:e21547
92. Nafee N, Husari A, Maurer CK, Lu C, de Rossi C, Steinbach
A etal (2014) Antibiotic-free nanotherapeutics: ultra-small,
mucus-penetrating solid lipid nanoparticles enhance the pul-
monary delivery and anti-virulence efficacy of novel quorum
sensing inhibitors. J Control Release: Off J Control Release
Society 192:131–140
93. Batoni G, Maisetta G, Brancatisano FL, Esin S, Campa M
(2011) Use of antimicrobial peptides against microbial bio-
films: advantages and limits. Curr Med Chem 18:256–279
94. Benito N, Franco M, Ribera A, Soriano A, Rodriguez-
Pardo D, Sorlí L etal (2016) Time trends in the aetiology
of prosthetic joint infections: a multicentre cohort study. Clin
Microbiol Infect: Off Publ Eur Soc Clin Microbiol Infect Dis
22(732):e1-8
95. Masters EA, Trombetta RP, de Mesy Bentley KL, Boyce BF,
Gill AL, Gill SR etal (2019) Evolving concepts in bone infec-
tion: redefining “biofilm”, “acute vs. chronic osteomyelitis”, “the
immune proteome” and “local antibiotic therapy.” Bone Res 7:20
96. Aguilera-Correa JJ, Gisbert-Garzarán M, Mediero A, Carias-
Cálix RA, Jiménez-Jiménez C, Esteban J etal (2022) Arabic
gum plus colistin coated moxifloxacin-loaded nanoparticles for
the treatment of bone infection caused by Escherichia coli. Acta
Biomater 137:218–237
97 Crane DP, Gromov K, Li D, Søballe K, Wahnes C, Büchner H
etal (2009) Efficacy of colistin-impregnated beads to prevent
multidrug-resistant A. baumannii implant-associated osteomy-
elitis. J Orthop Res: Off Publ Orthop Res Soc 27:1008–15
98. Aguilera-Correa JJ, Gisbert-Garzarán M, Mediero A, Fernández-
Aceñero MJ, de-Pablo-Velasco D, Lozano D etal (2022) Antibi-
otic delivery from bone-targeted mesoporous silica nanoparticles
for the treatment of osteomyelitis caused by methicillin-resistant
Staphylococcus aureus. Acta Biomaterialia 154:608–25
99. Birk SE, Mazzoni C, Mobasharah Javed M, Borre Hansen M,
Krogh Johansen H, Anders Juul Haagensen J etal (2021) Co-
delivery of ciprofloxacin and colistin using microcontainers for
bacterial biofilm treatment. Int J Pharm 599:120420
100. Alves D, Lopes H, Machado I, Pereira MO (2022) Colistin con-
ditioning surfaces combined with antimicrobial treatment to pre-
vent ventilator-associated infections. Biofouling 38:547–557
101. Pollini S, Boncompagni S, Di Maggio T, Di Pilato V, Spanu T,
Fiori B etal (2018) Invitro synergism of colistin in combination
with N-acetylcysteine against Acinetobacter baumannii grown
in planktonic phase and in biofilms. J Antimicrob Chemother
73:2388–2395
102. Shi Z, Puyo CA (2020) N-acetylcysteine to combat COVID-19:
an evidence review. Ther Clin Risk Manag 16:1047–1055
103. Aksoy N, Vatansever C, Zengin Ersoy G, Adakli Aksoy B, Fışgın
T (2022) The effect of biofilm inhibitor N-acetylcysteine on the
minimum inhibitory concentration of antibiotics used in Gram-
negative bacteria in the biofilm developed on catheters. Int J Artif
Organs 45:865–870
104. Ciacci N, Boncompagni S, Valzano F, Cariani L, Aliberti S, Blasi
F etal (2019) Invitro synergism of colistin and N-acetylcysteine
against Stenotrophomonas maltophilia. Antibiotics (Basel, Swit-
zerland) 8(3):101
105. Valzano F, Boncompagni SR, Micieli M, Di Maggio T, Di Pilato
V, Colombini L etal (2022) Activity of N-acetylcysteine alone
and in combination with colistin against Pseudomonas aerugi-
nosa biofilms and transcriptomic response to N-acetylcysteine
exposure. Microbiol Spectr 10:e0100622
106 Aiyer A, Das T, Whiteley GS, Glasbey T, Kriel FH, Farrell J
etal (2022) The efficacy of an N-acetylcysteine-antibiotic com-
bination therapy on Achromobacter xylosoxidans in a cystic
fibrosis sputum/lung cell model. Biomedicines 10(11):2886
107. Olofsson AC, Hermansson M, Elwing H (2003) N-acetyl-L-
cysteine affects growth, extracellular polysaccharide produc-
tion, and bacterial biofilm formation on solid surfaces. Appl
Environ Microbiol 69:4814–4822
108. Blasi F, Page C, Rossolini GM, Pallecchi L, Matera MG, Rogli-
ani P etal (2016) The effect of N-acetylcysteine on biofilms:
implications for the treatment of respiratory tract infections.
Respir Med 117:190–197
109. Doke SK, Raut JS, Dhawale S, Karuppayil SM (2014) Sensi-
tization of Candida albicans biofilms to fluconazole by terpe-
noids of plant origin. J Gen Appl Microbiol 60:163–168
110. Rangel MdL, Aquino SGd, Lima JMd, Castellano LR, Castro
RDd (2018) Invitro effect of Cinnamomum zeylanicum Blume
essential oil on Candida spp. involved in oral infections. Evid
Based Complement Alternat Med 2018:1–13
111. Shariati A, Didehdar M, Razavi S, Heidary M, Soroush F,
Chegini Z (2022) Natural compounds: a hopeful promise as
an antibiofilm agent against Candida species. Front Pharmacol
13:917787
112. Shinde RB, Raut JS, Chauhan NM, Karuppayil SM (2013)
Chloroquine sensitizes biofilms of Candida albicans to anti-
fungal azoles. Braz J Infect Dis 17:395–400
113. Guo T, Li M, Sun X, Wang Y, Yang L, Jiao H etal (2021)
Synergistic activity of capsaicin and colistin against colistin-
resistant Acinetobacter baumannii: invitro/vivo efficacy and
mode of action. Front Pharmacol 12:744494
114. Yao Z, Feng L, Zhao Y, Zhang X, Chen L, Wang L etal (2022)
Thymol increases sensitivity of clinical col-R Gram-negative
bacteria to colistin. Microbiol Spectr 10:e0018422
115. Zhou H, Xu M, Guo W, Yao Z, Du X, Chen L etal (2022) The
antibacterial activity of kaempferol combined with colistin
against colistin-resistant gram-negative bacteria. Microbiol
Spectr 10:e0226522
116. Wang Y, Kong J, Zhang X, Liu Y, Huang Z, Yuan L etal
(2022) Plumbagin resurrect colistin susceptible against colis-
tin-resistant Pseudomonas aeruginosa invitro and in vivo.
Front Microbiol 13:1020652
117. Xu M, Yao Z, Zhao Y, Shi S, Sun Y, Feng L etal (2022) Nar-
ingenin restores colistin activation against colistin-resistant
gram-negative bacteria invitro and invivo. Front Microbiol
13:916587
118 Topa SH, Palombo EA, Kingshott P, Blackall LL (2020) Activity
of cinnamaldehyde on quorum sensing and biofilm susceptibil-
ity to antibiotics in Pseudomonas aeruginosa. Microorganisms
8(3):455
119. Wang L, Zhang Y, Lin Y, Cao J, Xu C, Chen L, Zhou T (2023)
Resveratrol Increases Sensitivity of Clinical Colistin-Resistant
Pseudomonas aeruginosa to Colistin InVitro and InVivo. Micro-
biol Spectr 11(1):e01992–22
120. Zhao Y, Liu Y, Feng L, Xu M, Wen H, Yao Z etal (2022) Invitro
and invivo synergistic effect of chrysin in combination with
colistin against Acinetobacter baumannii. Front Microbiol
13:961498
121. LaSarre B, Federle MJ (2013) Exploiting quorum sensing to con-
fuse bacterial pathogens. Microbiol Mol Biol Rev 77:73–111
122. Pesci EC, Pearson JP, Seed PC, Iglewski BH (1997) Regulation
of las and rhl quorum sensing in Pseudomonas aeruginosa. J
Bacteriol 179:3127–3132
123. de Breij A, Gaddy J, van der Meer J, Koning R, Koster A,
van den Broek P etal (2009) CsuA/BABCDE-dependent pili
are not involved in the adherence of Acinetobacter baumannii
Brazilian Journal of Microbiology
1 3
ATCC19606(T) to human airway epithelial cells and their
inflammatory response. Res Microbiol 160:213–218
124. Lories B, Roberfroid S, Dieltjens L, De Coster D, Foster KR,
Steenackers HP (2020) Biofilm bacteria use stress responses to
detect and respond to competitors. Curr Biol: CB 30:1231–44.
e4
125. Ito A, May T, Kawata K, Okabe S (2008) Significance of rpoS
during maturation of Escherichia coli biofilms. Biotechnol Bio-
eng 99:1462–1471
126. Abdelaziz NA, Elkhatib WF, Sherif MM, Abourehab MAS,
Al-Rashood ST, Eldehna WM etal (2022) In silico docking,
resistance modulation and biofilm gene expression in multidrug-
resistant Acinetobacter baumannii via cinnamic and gallic acids.
Antibiotics (Basel, Switzerland) 11(7):870
127 Bellotti D, Remelli M (2022) Lights and shadows on the thera-
peutic use of antimicrobial peptides. Molecules (Basel, Switzer-
land) 27(14):4584
128. Rathinakumar R, Walkenhorst WF, Wimley WC (2009) Broad-
spectrum antimicrobial peptides by rational combinatorial design
and high-throughput screening: the importance of interfacial
activity. J Am Chem Soc 131:7609–7617
129. Raheem N, Straus SK (2019) Mechanisms of action for anti-
microbial peptides with antibacterial and antibiofilm functions.
Front Microbiol 10:2866
130. Morroni G, Sante LD, Simonetti O, Brescini L, Kamysz W,
Kamysz E etal (2021) Synergistic effect of antimicrobial pep-
tide LL-37 and colistin combination against multidrug-resistant
Escherichia coli isolates. Future Microbiol 16:221–227
131. Dosler S, Karaaslan E (2014) Inhibition and destruction of Pseu-
domonas aeruginosa biofilms by antibiotics and antimicrobial
peptides. Peptides 62:32–37
132. Zhang Y, He X, Cheng P, Li X, Wang S, Xiong J etal (2021)
Effects of a novel anti-biofilm peptide CRAMP combined with
antibiotics on the formation of Pseudomonas aeruginosa bio-
films. Microb Pathog 152:104660
133. Jorge P, Grzywacz D, Kamysz W, Lourenço A, Pereira MO
(2017) Searching for new strategies against biofilm infections:
colistin-AMP combinations against Pseudomonas aeruginosa and
Staphylococcus aureus single- and double-species biofilms. PLoS
One 12:e0174654
134. Bardbari AM, Arabestani MR, Karami M, Keramat F, Aghaza-
deh H, Alikhani MY etal (2018) Highly synergistic activity of
melittin with imipenem and colistin in biofilm inhibition against
multidrug-resistant strong biofilm producer strains of Acineto-
bacter baumannii. Eur J Clin Microbiol Infect Dis: Off Publ Eur
Soc Clin Microbiol 37:443–454
135. Mahlapuu M, Håkansson J, Ringstad L, Björn C (2016) Anti-
microbial peptides: an emerging category of therapeutic agents.
Front Cell Infect Microbiol 6:194
136. Luo J, Dong B, Wang K, Cai S, Liu T, Cheng X etal (2017)
Baicalin inhibits biofilm formation, attenuates the quorum sens-
ing-controlled virulence and enhances Pseudomonas aeruginosa
clearance in a mouse peritoneal implant infection model. PLoS
One 12:e0176883
137. Dai L, Wu TQ, Xiong YS, Ni HB, Ding Y, Zhang WC etal
(2019) Ibuprofen-mediated potential inhibition of biofilm
development and quorum sensing in Pseudomonas aeruginosa.
Life Sci 237:116947
138. Chegini Z, Khoshbayan A, Vesal S, Moradabadi A, Hashemi A,
Shariati A (2021) Bacteriophage therapy for inhibition of multi
drug-resistant uropathogenic bacteria: a narrative review. Ann
Clin Microbiol Antimicrob 20:30
139. Vashisth M, Yashveer S, Jaglan AB, Virmani N, Bera BC, Vaid
RK etal (2022) Synergy of a virulent phage (φAB182) with
antibiotics leading to successful elimination of biofilms formed
by MDR Acinetobacter baumannii. Can J Microbiol 68:731–746
140. Soontarach R, Nwabor OF, Voravuthikunchai SP (2022) Interac-
tion of lytic phage T1245 with antibiotics for enhancement of
antibacterial and anti-biofilm efficacy against multidrug-resistant
Acinetobacter baumannii. Biofouling 38:994–1005
141. Wintachai P, Phaonakrop N, Roytrakul S, Naknaen A, Pomwised
R, Voravuthikunchai SP etal (2022) Enhanced antibacterial
effect of a novel Friunavirus phage vWU2001 in combination
with colistin against carbapenem-resistant Acinetobacter bau-
mannii. Sci Rep 12:2633
142. Jansen M, Wahida A, Latz S, Krüttgen A, Häfner H, Buhl EM
etal (2018) Enhanced antibacterial effect of the novel T4-like
bacteriophage KARL-1 in combination with antibiotics against
multi-drug resistant Acinetobacter baumannii. Sci Rep 8:14140
143. Comeau AM, Tétart F, Trojet SN, Prère MF, Krisch HM (2007)
Phage-antibiotic synergy (PAS): beta-lactam and quinolone anti-
biotics stimulate virulent phage growth. PLoS One 2:e799
144. Kamal F, Dennis JJ (2015) Burkholderia cepacia complex phage-
antibiotic synergy (PAS): antibiotics stimulate lytic phage activ-
ity. Appl Environ Microbiol 81:1132–1138
145. khilonawala F, Majeed Ali A, AL-Shaheen MR (2023) Elec-
trospun Fibers with Lactobacillus Acidophilus: A Poten- tial
InVitro Solution Against Gardnerella Infections. Trends Pharm
Biotechnol 1(1):1–11. https:// doi. org/ 10. 57238/ tpb. 2023. 144236.
1000
146 Baliga P, Goolappa PT, Shekar M, Kallappa GS (2022) Cloning,
characterization, and antibacterial properties of endolysin LysE
against planktonic cells and biofilms of Aeromonas hydrophila.
Probiotics Antimicrob Proteins 15(3):646–654
147. Hanlon GW (2007) Bacteriophages: an appraisal of their role
in the treatment of bacterial infections. Int J Antimicrob Agents
30:118–128
148. Chen X, Liu M, Zhang P, Xu M, Yuan W, Bian L etal (2022)
Phage-derived depolymerase as an antibiotic adjuvant against
multidrug-resistant Acinetobacter baumannii. Front Microbiol
13:845500
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
... 6 Additionally, monotherapy could induce resistance through the increased formation of biofilms, such as colistin. 7 Thus, PA biofilm infections are difficult to treat. ...
Article
Full-text available
Objective Pseudomonas aeruginosa (PA) often displays drug resistance and biofilm-mediated adaptability. Here, we aimed to evaluate the antibiofilm efficacy of azithromycin-based combination regimens. Methods Minimum inhibitory concentrations (MICs), minimal biofilm eradication concentrations (MBECs), and MBEC-combination of azithromycin, colistin, amikacin, and levofloxacin to bioluminescent strain PAO1 and carbapenem-resistant PAO1 (CRPAO1) were assessed. An animal biofilm infection model was established and detected using a live animal bio-photonic imaging system. Results In vitro, PAO1 and CRPAO1 were susceptible to colistin, amikacin, and levofloxacin, while they were unsusceptible to azithromycin. The combinations based on azithromycin have no synergistic effect on biofilm in vitro. In vivo, azithromycin plus colistin or levofloxacin could shorten the PAO1 biofilm eradication time, which totally eradicates the biofilm in all mice on the 8th or 6th day, while monotherapy only eradicate biofilm in 70% or 80% mice on the 8th day. For CRPAO1 biofilm, only azithromycin–colistin combination and colistin monotherapy eradicated the bacteria in 60% and 40% of mice at the 6th day. Conclusion Azithromycin-based combinations containing levofloxacin or colistin had no synergistic effect in vitro, and they are promising for clinical applications due to the good synergistic activity against PAO1 biofilms in vivo.
Thesis
Full-text available
Staphylococcus aureus is an important human pathogen that causes a wide variety of life-threatening infections ranging from minor skin and oral infections to severe infections, such as bacteremia, pneumonia, osteomyelitis, or endocarditis due to the presence and secretion of a large number of virulence factors that are controlled by global virulence regulators in complex networks. Furthermore, S. aureus infections have become a threat to public health because of their high potential to form biofilm, and their ability to resist a wide range of antibiotics has exacerbated further. Therefore, understanding the regulatory networks and developing a drug targeting these networks has the potential to stand as therapeutic targets for future treatment of antibiotic resistant infections. In a previous study msaC was identified as the modulator of sarA, a new global virulence regulator that controls the expression of sarA and biofilm development. Furthermore, it has also been shown that msaC is a part of four-gene operon, msaABCR operon, which includes four-genes: SAUSA300_1296 (msaA), SAUSA300_1295 (msaB), SAUSA300_1294 (msaC), and antisense RNA, msaR. The mechanism of regulation of msaABCR operon and the function of individual genes were not clearly known yet. This study defines the role of msaB, the second gene of the msaABCR operon, which will help shed some light on the regulation of msaABCR. We deleted msaB gene from USA300_LAC, and studied the major msaB phenotypes: pigmentation, protease production, biofilm formation, and rate of cell death. Deletion of msaB
Article
Full-text available
Peatlands are important sources of the greenhouse gas methane emissions equipoised by methanogens and methanotrophs. However, knowledge about how microbial functional groups associated with methane production and oxidation respond to water table fluctuations has been limited to date. Here, methane-related microbial communities and the potentials of methane production and oxidation were determined along sectioned peat layers in a subalpine peatland across four Sphagnum -dominated sites with different water table levels. Methane fluxes were also monitored at these sites. The results showed that mcrA gene copies for methanogens were the highest in the 10- to 15-cm peat layer, which was also characterized by the maximum potential methane production (24.53 ± 1.83 nmol/g/h). Copy numbers of the pmoA gene for type Ia and Ib methanotrophs were enriched in the 0–5 cm peat layer with the highest potential methane oxidation (43.09 ± 3.44 nmol/g/h). For the type II methanotrophs, the pmoA gene copies were higher in the 10- to 15-cm peat layer. Hydrogenotrophic methanogens and type II methanotrophs dominated the methane functional groups. Deterministic process contributed more to methanogenic and methanotrophic community assemblages in comparison with stochastic process. The level of water table significantly shaped methanogenic and methanotrophic community structures and regulated methane fluxes. Compared with vascular plants, Sphagnum mosses significantly reduced the methane emissions in peatlands. Collectively, these findings enhance a comprehensive understanding of the effect of the water table level on methane functional groups, with consequential implications for reducing methane emissions within peatland ecosystems. IMPORTANCE The water table level is recognized as a critical factor in regulating methane emissions, which are largely dependent on the balance of methanogens and methanotrophs. Previous studies on peat methane emissions have been mostly focused on spatial-temporal variations and the relationship with meteorological conditions. However, the role of the water table level in methane emissions remains unknown. In this work, four representative microhabitats along a water table gradient in a Sphagnum -dominated peatland were sampled to gain an insight into methane functional communities and methane emissions as affected by the water table level. The changes in methane-related microbial community structure and assembly were used to characterize the response to the water table level. This study improves the understanding of the changes in methane-related microbial communities and methane emissions with water table levels in peatlands.
Article
Full-text available
Colistin resistance is a globalized sensible issue because it has been considered a drug of the last-line resort to treat drug-resistant bacterial infections. The product of the mobilized colistin resistance (mcr) gene and its variants are the significant causes of colistin resistance, which is emerging due to the frequent colistin use in veterinary, and these genes circulate among the bacterial community. Apart from mcr genes, some other intrinsic genes and proteins are also involved in colistin resistance. Researchers focus on the most advanced genomics (whole genome sequencing), proteomics, and bioinformatics approaches to explore the question of colistin resistance. To combat colistin resistance, researchers developed various strategies such as the development of newer drugs, the repurposing of existing drugs, combinatorial treatment by colistin with other drugs, a nano-based approach, photodynamic therapy, a CRISPRi-based strategy, and a phage-based strategy. In this timeline review, we have discussed the development of colistin resistance and its management in developing countries.
Article
Full-text available
Infections caused by colistin-resistant P. aeruginosa strains pose a serious threat to public health. It is therefore urgent to find new strategies to deal with these bacterial infections. We aimed to investigate the efficacy and mechanisms of the colistin/resveratrol combination in eradicating colistin-resistant P. aeruginosa isolates and their biofilms both in vitro and in vivo. The results revealed that six clinically isolated colistin-resistant P. aeruginosa strains were multidrug resistant (MDR) strains, and resveratrol showed no antimicrobial activity against eight P. aeruginosa strains. Checkerboard assay and time-kill assays indicated that the combination therapy of resveratrol and colistin indicated a remarkable synergistic effect in vitro, and biofilm assays and SEM indicated synergistic antibiofilm activity. Furthermore, this combination could efficiently eliminate MDR bacteria in a murine infection model and improve the survival rate of Galleria mellonella. Fluorescence analysis, ALP, and β-galactosidase activity test results indicated that the colistin/resveratrol combination increased the membrane permeability of bacteria. In conclusion, our results may provide an efficient alternative pathway against colistin-resistant P. aeruginosa infections. IMPORTANCE P. aeruginosa is a ubiquitous Gram-negative opportunistic pathogen associated with a wide array of life-threatening acute and chronic infections. However, the improper and excessive use of antibiotics has contributed to the increasing emergence of multidrug-resistant (MDR) P. aeruginosa, even colistin-resistant strains, which presents a major challenge to clinical anti-infection treatment. Resveratrol, a naturally occurring polyphenolic antioxidant, can effectively slow down or avoid the occurrence and development of bacterial resistance and is expected to offer a promising strategy to overcome bacterial infections. In this study, colistin/resveratrol combination could synergistically damage the bacterial cell membrane, thereby inducing cell lysis while addressing the emergence of drug resistance. Moreover, this combination therapy may provide an efficient alternative pathway to combat the colistin-resistant P. aeruginosa in clinical practice.
Article
Full-text available
Haemophilus influenzae is a pathogen that causes invasive bacterial infections in humans. The highest prevalence lies in both young children and adults. Generally, there are no vaccines available that target all the strains of Haemophilus influenzae. Hence, the purpose of this research is to employ bioinformatics and immunoinformatics approaches to design a Multi-Epitope Vaccine candidate employing the pathogenic cell division protein FtsN that specifically combat all the Haemophilus influenzae strains. The current research focuses on developing subunit vaccine in contrast to vaccines generated from the entire pathogen. This will be accomplished by combining multiple bioinformatics and immunoinformatics approaches. As a result, prospective T cells (helper T lymphocyte and cytotoxic T lymphocytes) and B cells epitopes were investigated. The human leukocyte antigen allele having strong associations with the antigenic and overlapping epitopes were chosen, with 70% of the total coverage of the world population. To construct a linked vaccine design, multiple linkers were used. To increase the immunogenic profile, an adjuvant was linked using EAAAK linker. The final vaccine construct with 149 amino acids was obtained after adjuvants and linkers were added. The developed Multi-Epitope Vaccine has a high antigenicity as well as viable physiochemical features. The 3D conformation was modeled and undergoes refinement and validation using bioinformatics methods. Furthermore, protein–protein molecular docking analysis was performed to predict the effective binding poses of Multi-Epitope Vaccine with the Toll-like receptor 4 protein. Besides, vaccine underwent the codon translational optimization and computational cloning to verify the reliability and proper Multi-Epitope Vaccine expression. In addition, it is necessary to conduct experiments and research in the laboratory to demonstrate that the vaccine that has been developed is immunogenic and protective.
Article
Full-text available
Cystic fibrosis (CF) is a disorder causing dysfunctional ion transport resulting in the accumulation of viscous mucus. This environment fosters a chronic bacterial biofilm-associated infection in the airways. Achromobacter xylosoxidans, a gram-negative aerobic bacillus, has been increasingly associated with antibiotic resistance and chronic colonisation in CF. In this study, we aimed to create a reproducible model of CF infection using an artificial sputum medium (ASMDM-1) with bronchial (BEAS-2B) and macrophage (THP-1) cells to test A. xylosoxidans infection and treatment toxicity. This study was conducted in three distinct stages. First, the tolerance of BEAS-2B cell lines and two A. xylosoxidans strains against ASMDM-1 was optimised. Secondly, the cytotoxicity of combined therapy (CT) comprising N-acetylcysteine (NAC) and the antibiotics colistin or ciprofloxacin was tested on cells alone in the sputum model in both BEAS-2B and THP-1 cells. Third, the efficacy of CT was assessed in the context of a bacterial infection within the live cell/sputum model. We found that a model using 20% ASMDM-1 in both cell populations tolerated a colistin–NAC-based CT and could significantly reduce bacterial loads in vitro (~2 log10 CFU/mL compared to untreated controls). This pilot study provides the foundation to study other bacterial opportunists that infect the CF lung to observe infection and CT kinetics. This model also acts as a springboard for more complex co-culture models.
Article
Full-text available
Social hygiene is seriously threatened by the rise in colistin (COL) resistance against Gram-negative bacteria (GNB). With resistance to last-line antibiotics such as COL becoming more common, it is imperative to identify alternative treatment options. In our work, we sought to determine if COL plus kaempferol (KP) present synergistic effects on the antibacterial and antibiofilm activities against colistin-resistant (Col-R) GNB in vivo and in vitro. Twenty-four Col-R GNB were collected as the experimental strains. The synergistic activity of COL and KP was evaluated by checkerboard method, time-killing assays, and the Galleria mellonella experiment. The antibiofilm effectiveness of the COL/KP combination against Col-R GNB was assessed using biofilm inhibition and eradication assays and scanning electron microscopy (SEM). Cytotoxicity tests were performed to detect the toxicity of KP monotherapy or combination therapy. There is synergistic antibacterial activity of COL and KP combination in vitro. KP combined with COL could inhibit the formation of bacterial biofilms. The amalgamation of COL and KP considerably reduced the amount of bacteria in the biofilm, according to the SEM findings. The COL/KP combination improved the survivorship of infected larvae in the G. mellonella in vivo infection model. In addition, the combination of KP and COL showed no cytotoxicity at synergistic combined concentrations according to cytotoxicity assays. This represents the first account of the antibacterial and antibiofilm activities of KP in combination with COL against Col-R GNB. Therefore, our results may provide an effective alternative route to combat Col-R GNB infections. IMPORTANCE COL is one of the few antibiotics effective against clinical isolates of GNB. However, in recent years, GNB resistance to colistin has been increasing. As a result, the combined application of colistin in conjunction with nonantibacterial medications has garnered considerable interest. In this work, the KP/COL combination showed effective antibacterial and antibiofilm activities in vitro and in vivo. The synergistic effect of combined application may be attributed to membrane permeability. Due to the low cytotoxicity of the combined concentration, the combination exhibits a promising future for use in clinical anti-infection treatments. This finding might broaden the potential applications for COL.
Article
Biofilms associated with multidrug-resistant (MDR) Acinetobacter baumannii on medical devices remain a big clinical problem. Antibiotic susceptibility tests were performed with eight commonly employed antibiotics against clinical isolates. The effects of antibiotics in combination with well-characterized lytic phage T1245 were studied to assess their antibacterial and anti-biofilm efficacy. Ceftazidime, colistin, imipenem, and meropenem significantly reduced bacterial density up to approximately 80% when combined with phage T1245, compared with control. Phage T1245 in combination with ceftazidime, colistin, and meropenem at subinhibitory concentrations demonstrated significant reduction in biomass and bacterial viability of 3-day established biofilms, compared with antibiotic alone. In addition, electron microscopy further confirmed the disruption of biofilm structure and cell morphology upon treatment with phage T1245 and antibiotics, including ceftazidime, colistin, and meropenem. Combined treatment of phage T1245 with these antibiotics could be employed for the management of A. baumannii infections and eradication of the bacterial biofilms.
Article
Osteomyelitis is a hard-to-treat infection of the bone and bone marrow that is mainly caused by Staphylococcus aureus, with an increasing incidence of methicillin-resistant S. aureus (MRSA). Owing to the aggressiveness of these bacteria in colonizing and destroying the bone, systemic antibiotic treatments fail to eradicate the infection. Instead, it normally entails surgery to remove the dead or infected bone. In this work, we report bone-targeted mesoporous silica nanoparticles for the treatment of osteomyelitis. The nanoparticles have been engineered with a functional gelatine/colistin coating able to hamper premature release from the mesopores while effectively disaggregating the bacterial biofilm. Because antibiotic resistance is a global emergency, we have designed two sets of identical nanoparticles, carrying each of them a clinically relevant antibiotic, that have demonstrated to have synergistic effect. The bone-targeted nanoparticles have been thoroughly evaluated in vitro and in vivo, obtaining a notable reduction of the amount of bacteria in the bone in just 24 h after only one dose, and paving the way for localized, nanoparticle-mediated treatment of MRSA-caused osteomyelitis. Statement of significance In this work, we propose the use of bone-targeted mesoporous silica nanoparticles to address S. aureus-caused osteomyelitis that render synergistic therapeutic effect via multidrug delivery. Because the bacterial biofilm is responsible for an aggressive surgical approach and prolonged antibiotic treatment, the nanoparticles have been functionalized with a functional coating able to both disaggregate the biofilm, hamper premature antibiotic release and protect the intact bone. These engineered nanoparticles are able to effectively target bone tissue both in vitro and in vivo, showing high biocompatibility and elevated antibacterial effect.