You are on page 1of 12

Microbial Pathogenesis 142 (2020) 104050

Contents lists available at ScienceDirect

Microbial Pathogenesis
journal homepage: www.elsevier.com/locate/micpath

Combinations of early generation antibiotics and antimicrobial peptides are T


effective against a broad spectrum of bacterial biothreat agents
Christopher K. Cotea,∗, Irma I. Blancoa, Melissa Huntera, Jennifer L. Shoea,
Christopher P. Klimkoa, Rekha G. Panchalb, Susan L. Welkosa,∗∗
a
Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702-5011,
USA
b
Countermeasures Division, USAMRIID, Fort Detrick, MD, USA

ARTICLE INFO ABSTRACT

Keywords: The misuse of infectious disease pathogens as agents of deliberate attack on civilians and military personnel is a
Antimicrobial peptides serious national security concern, which is exacerbated by the emergence of natural or genetically engineered
Antibiotics multidrug resistant strains. In this study, the therapeutic potential of combinations of an antibiotic and a broad-
Resistance spectrum antimicrobial peptide (AMP) was evaluated against five bacterial biothreats, the etiologic agents of
Bioterror agents
glanders (Burkholderia mallei), melioidosis (Burkholderia pseudomallei), plague (Yersinia pestis), tularemia
Therapeutics
(Francisella tularensis), and anthrax (Bacillus anthracis). The therapeutics included licensed early generation
Combinations
antibiotics which are now rarely used. Three antibiotics and one 24- amino acid AMP were selected based on
MIC assay data. Combinations of the AMP and tigecycline, minocycline, or novobiocin were screened for sy-
nergistic activity by checkerboard MIC assay. The combinations each enhanced the susceptibility of several
strains. The tetracycline-peptide combinations increased the sensitivities of Y. pestis, F. tularensis, B. anthracis and
B. pseudomallei, and the novobiocin-AMP combination augmented the sensitivity of all five. In time-kill assays,
down-selected combinations of the peptide and minocycline or tigecycline enhanced killing of B. anthracis, Y.
pestis, F. tularensis, and Burkholderia mallei but not B. pseudomallei. The novobiocin-AMP pair significantly re-
duced viability of all strains except B. mallei, which was very sensitive to the antibiotic alone. The results
suggested that antibiotic-AMP combinations are useful tools for combating diverse pathogens. Future studies
employing cell culture and animal models will utilize virulent strains of the agents to investigate the in vivo
availability, host cytotoxicity, and protective efficacy of these therapeutics.

1. Introduction especially hazardous and resistant bacteria. These therapeutic options


would include early generation licensed antibiotics which are now
The emergence of bacteria which are resistant to antibiotics has rarely used due to their perceived excessive toxicity and limited ther-
been an increasing problem in treating bacterial infections. This is an apeutic window.
especially critical concern for biothreat pathogens as some of these A second strategy utilizes antimicrobial peptides (AMPs), a diverse
strains already have high natural resistance to a range of antimicrobials class of antimicrobials which are naturally occurring or synthetically
(e.g., the Burkholderia). Furthermore, the development of multiple an- produced, are 12–50 amino acids in length, and exhibit broad-spectrum
tibiotic resistant (AMR) remains a possible catastrophic scenario. Thus, activity [1,2]. Although bacterial resistance to AMPs has been reported,
the development of new antimicrobial strategies is an urgent need. In it is considered to be rare [2,3]. Thus AMPs are promising non-anti-
one strategy, previously characterized but overlooked antimicrobials biotic accessory therapeutics [1,2,4]. They include well-studied natu-
could be repurposed as potentially successful new treatments for rally occurring AMPs, e.g., cecropin A, LL-37, magainin II, mastoparan,

Abbreviations: AMP, antimicrobial peptide; Bm, Burkholderia mallei; Bp, Burkholderia pseudomallei; Yp, Y. pestis; Ft, F. tularensis; Ba, B. anthracis; Ec, Escherichia coli;
Ps, Pseudomonas aeruginosa; LBG, Luria-Bertani medium with glycerol; MIC, antimicrobial minimal inhibitory concentration; MBC, minimum bactericidal con-
centration; FICI, Fractional Inhibitory Concentration Index; combo, combination; AMR, antimicrobial resistant; combo, combination

Corresponding author.
∗∗
Corresponding author.
E-mail addresses: christopher.k.cote.civ@mail.mil (C.K. Cote), susan.l.welkos.vol@mail.mil (S.L. Welkos).

https://doi.org/10.1016/j.micpath.2020.104050
Received 4 December 2019; Received in revised form 6 February 2020; Accepted 7 February 2020
Available online 09 February 2020
0882-4010/ Published by Elsevier Ltd.
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

and melittin [5–13]. Also, synthetic peptides have been engineered In summary, the presence of AMR strains of biothreat bacteria can
based on natural AMPs. For example, the 12 amino acid cationic pep- potentially limit the availability of effective therapeutics. Treatment
tide bactenecin inhibits growth of some gram-negative bacteria. Wu options may be greatly limited, especially with Burkholderia infections
et al. [14] constructed several bactenecin analogs that exhibited in- of which recurrent infections are also a significant problem. Thus, the
creased inhibitory activity for both gram-negative and gram-positive development of new antimicrobial therapies, such as the proposed
bacteria [14,15]. Hybrid AMPs have also been designed based on ce- combination strategy, is critically important. Furthermore, as shown for
cropin A, LL-37, and magainin II and have been shown to provide other novel therapeutic candidates, the proposed combination ap-
broad-spectrum antimicrobial activity [9]. AMPs have long been of proach may allow the development of more flexible, patient- and in-
interest as potentially clinically relevant therapeutics. Several limita- fection-specific options for customizing treatment.
tions have delayed their successful clinical use. Natural AMPs can be The goal of our studies is to identify antibiotics which, when com-
inhibited by their sensitivity to adverse host-like conditions (e.g., acidic bined with an AMP, exhibit enhanced antibacterial activity against a
pH, salts, protease digestion); and nonspecific binding to off-target host broad spectrum of bacterial biothreats and their AMR variants. The
tissues and serum proteins. Also, their use is hampered by potential host antibiotics selected included licensed, early generation antibiotics
toxicity [1,2,16,17]. However, these limitations may be significantly which are currently rarely used and have the potential to be effective
ameliorated by use of engineered peptides such as WLBU2, a cationic against five bacterial biothreat agents: the etiologic agents of glanders
helical peptide, which was optimized in length and amino acid content (Burkholderia mallei), melioidosis (Burkholderia pseudomallei), plague
for antibacterial activity. It was reported to have in vitro activity against (Yersinia pestis), tularemia (Francisella tularensis), and anthrax (Bacillus
three gram-negative biothreat strains; kill intracellular pathogens in anthracis). Due to their extensive resistance to many AMPs and innate
mouse and human cells; and prevent biofilm development on epithelial antibiotic resistance, the Burkholderia spp. were used as the “bench-
cells, with little or no cytotoxicity [4,18–20]. Finally, bacteriocins re- mark” for assessing AMP and antibiotic sensitivity. In future work,
present another class of AMPs that could be repurposed to treat bio- down-selected antibiotic-AMP combinations with synergistic or en-
threat agents. They specifically destabilize the plasma membranes of hanced in vitro activity will be evaluated for their pharmacokinetic
bacteria, causing cell lysis, and thus are widely regarded to be safe for properties, safety, and therapeutic efficacy in animal models.
humans [21,22].
Naghmouchi et al. [23] showed that there are synergistic effects 2. Materials and Methods
between the early generation antibiotic colistin and bacteriocin when
used in combination against gram-negative pathogens such as Pseudo- 2.1. Bacterial strains, media, and growth conditions
monas aeruginosa (Pa). Until recently, colistin had fallen out of favor
and was only used as a last resort for AMR gram-negative bacteria The bacterial strains used included five exempt select-agent strains
because of its potential nephrotoxicity. However, when colistin was derived from the etiologic agents of glanders, melioidosis, plague, tu-
combined with a bacteriocin the efficacy of both increased, thus low- laremia, and anthrax: B. mallei (Bm), B. pseudomallei (Bp), Y. pestis (Yp),
ering their effective concentrations and reducing toxicity of the com- F. tularensis (Ft), and B. anthracis (Ba), respectively. These strains are
bination for mammalian cells [23]. Since resistance to colistin has re- attenuated mutants derived from a wild type prototype strain as de-
cently emerged, the reduction in the antibiotic's effective dose afforded scribed in Table 1 [32–37]. Suspensions of the bacteria were freshly
by a synergistic therapeutic combination is especially vital. prepared for assays using strain stocks maintained at −80 °C. Strains Bp
B. pseudomallei, the agent of melioidosis, and B. mallei, responsible JW270 and E. coli (Ec) ATCC 25922 (quality control strain) were cul-
for glanders, are considered to be potential biological warfare or bio- tured on sheep blood agar plates (SBAP, Thermo Fisher-Remel) and
terrorism agents and are listed as a Tier 1 biological select agent by the incubated at 37 °C for 24 h. Bm CLH001 was grown on Luria-Bertani
US Department of Health and Human Services [24] (www.selectagents. medium (Lennox formulation) with 4% glycerol (LBG) agar plates
gov/selectagentsandtoxinslist.html). They are intrinsically resistant to supplemented with 200 μM iron sulfate and incubated at 37 °C for 48 h
many antibiotics, including beta-lactam antibiotics, aminoglycosides, [32]. Yp pgm- pPst− was streaked on an SBAP and grown for 48 h at
macrolides, and polymixins such as colistin. However other older, 28–30 °C. Ft LVS was cultured on a chocolate agar (CA) plate (Thermo
rarely used antimicrobials besides colistin are available to potentially Fisher-Remel) for 48 h at 37 °C, colonies were suspended in cation-
counter drug resistant bacteria such as the Burkholderia, e.g., fosfo- adjusted Mueller-Hinton Broth (MHB) (BBL™, BD Diagnostics Franklin
mycin, nitrofurantoin, tigecycline, minocycline, fusidic acid, and others Lake, NJ), and the suspension used to inoculate a flask with 10 ml MHB
[25–30]. For instance, novobiocin is a narrow-spectrum antibiotic supplemented with 2% IsovitaleX (BBL™, BD Diagnostics) from a freshly
considered to be primarily effective against gram-positive pathogens rehydrated vial. The broth culture was incubated 24 h at 37 °C, with
but less effective for treating infections caused by gram-negative bac- shaking at 200 rpm and adjusted to the required concentration. Stocks
teria due to reduced permeability or their outer membrane. However of purified ungerminated spores of Ba strain Sterne were prepared as
evidence supports the use of various small molecules which can described previously [38,39]. The spores were incubated in germinant
permeate membranes to facilitate the concomitant entry of antibiotics consisting of 50 mM L-alanine and 100 mM inosine for 25 min at 37 °C,
such as novobiocin [30,31]. Furthermore, these antibiotic adjuncts may at which time the spores had uniformly germinated as shown by their
effectively reduce the antibiotic minimal inhibitory concentration loss of refractility by phase microscopy [38,39]. The spore suspension
(MIC), and we hypothesize that a similar phenomenon could occur by was then diluted in buffer or MHB and adjusted to the concentration
combining an AMP with an antibiotic. required for the assay.

Table 1
Panel of surrogate bacterial strains used.
Bacterial strain (abbreviation)a Characteristics Source References

B. anthracis Sterne (Ba) capsule-negative strain; animal vaccine USAMRIID 33


Y. pestis pgm− pPst− (Yp) pgm- pPst- mutant of Yp strain CO92 USAMRIID 36, 37
B. pseudomallei JW270 (Bp) capsule-negative mutant of Bp strain DD503 J. Warawa, NIAID 34
B. mallei CLH001 ΔtonB, Δhcp1 (Bm) siderophore and virulence gene deletion mutant of Bm ATCC23344 A. Torres, UTMC 32
F. tularensis subsp holartica LVS (Ft) spontaneous attenuated derivative; live vaccine strain USAMRIID 35

a
Highly attenuated and select agent-excluded strains derived from prototype select agent pathogenic strains.

2
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

2.2. Chemicals, antibiotics and peptides yield the appropriate concentration (1.5–5 × 105/ml) in 100 μl MHB.
The plates were incubated for 24 h at 37 °C (or as indicated in 2.3.1) in
Chemicals were obtained from Sigma-Aldrich (St. Louis, MO) except ambient air. One well with no antibiotic was used as a positive growth
as indicated. Antibiotics were procured from Sigma-Aldrich except for control on each plate. Plates were read for visual turbidity to determine
fosfomycin and ceftazidime, which were obtained from USP. the MIC. The antbiotics tested included nalidixic acid, colistin, cefta-
Antimicrobial peptides were acquired from the following sources: zidime, tigecycline, minocycline, and novobiocin; and the AMPs tested
Sigma/Fluka, Bachem (Torrance, CA), Biopeptek (Malvern, PA), and A included nisin and WLBU2. These data are representative of at least two
&A Labs/Synthetic Biomolecules (San Diego, CA). They included separate experiments of similar results per strain. The MIC data were
WLBU2, BMAP-18, mastoparan7, nisin, melittin, magaininII, bacte- evaluated for synergistic effects of the combinations by determining
nicin, and CAMA, and their synthesis and activity have been previously Fractional Inhibitory Concentration Index (FICI) values in accordance
reported [5,7,9,15,20,23]. Stocks of antibiotics and AMPs were pre- with the following formula: The FICI = FICA + FICB, where
pared in concentrations of 5–10 mg/ml in accordance with manu- FICA = MIC of drug A in combination/MIC of drug A alone, and
facturer recommendations and stored in single use aliquots at −80 °C. FICB = MIC of drug B in combination/MIC of drug B alone. The FICIs
are then interpreted using these breakpoints for four categories: Sy-
2.3. Antimicrobial susceptibility tests nergy: FICI of <0.5; Additivity: FICI of >0.5 to <1; no interaction
(Indifference): FICI of >1 to <4; and Antagonism: FICI of >4
2.3.1. Standard assay [45,47–49].
In vitro susceptibility assays of antimicrobial MIC were performed in
accordance with Clinical & Laboratory Standards Institute (CLSI) stan- 2.4. Direct killing assays
dards in 96-well microtiter plates with cation-adjusted MHB.
Antimicrobial stock solutions, bacterial inocula, and test conditions In vitro assays of direct antimicrobial activity were performed in the
were as described previously [40–43]. Instead of using a vegetative cell absence of bacterial growth according to a modification of the proce-
suspension, Ba Sterne inocula were prepared from freshly germinated dure described by Abdelbaqi et al. [18]. Dilutions of selected anti-
spores as specified above. The bacterial inocula were prepared from microbial agents, alone or combined, were prepared and added in vo-
suspensions of freshly grown plated colonies, or from an overnight lumes of 100 μl to wells in triplicate on a 96-well microtiter plate. A
broth culture (Ft LVS) and adjusted to a final concentration based on the suspension of bacteria was made and adjusted by OD600 in a non-nu-
OD600 reading of 1.5–5 × 105 CFU/ml. Positive growth control wells trient containing buffer to a concentration producing 1.5–5 × 105 CFU
included bacteria incubated in buffer without antimicrobial agent. For per well (in a final volume of 200 μl). Positive growth control wells
assays employing antimicrobial stocks prepared in DMSO, the buffer included the bacteria incubated in buffer without antimicrobial agent.
used in the control wells contained DMSO at a concentration equal to In all assays employing antibiotic/AMP stocks prepared with DMSO, the
that of the highest concentration of DMSO in the antimicrobial-treated latter was added to the control wells as described in 2.3.1. The in-
samples to account for any residual antimicrobial activity of DMSO. oculated plates were incubated for 2 h at the appropriate temperature
MICs were determined by titration dilution covering a broad range of and serial dilutions then plated in triplicate on solid medium for viable
antimicrobial concentration, typically 0.0312–62.5 μg/ml except as count determinations. Survival was assessed by comparing the change
indicated. After inoculation with the bacteria, the plates were incubated in counts of AMP/antibiotic-treated compared to buffer-treated con-
at 37 °C, except for Yp which was incubated at 28 °C [41]. Yp modifies trols. The buffer used for preparing the assay plate and diluting samples
its surface LPS structure in various ways, e.g., by reducing the extent of for plating was Gibco PBS (Thermo Fisher Scientific, Waltham, MA,
lipid A acylation, in response to the change in temperature from ≤27 °C USA) for all strains except Yp, for which a buffer with 10 mM potassium
to 35–37 °C. This change may potentially alter the activity or interac- phosphate pH 7.4 was prepared and used.
tion of AMPs with the bacterium, a possibility which was not evaluated
in the current study. However, in the current and previous studies, 2.5. Time kill assays
outer-membrane active antibiotics and AMPs have been shown to be
active in vitro and in vivo, alone or in antibiotic-peptide combinations, Time-associated in vitro killing assays were performed and evaluated
against gram-negative bacteria to include Yp, as will be described in the by a modification of previously described methods [43,45,46,50]. In
Results and Discussion. In addition, Yp grows faster at 28 °C than at brief, flasks containing MHB, with or without addition of an anti-
37 °C, and the three virulence plasmids of Yp are maintained more microbial(s), were inoculated with a concentration of bacteria adjusted
stably at 28 °C than at 37 °C [41]. Furthermore, in MIC screens with as described for the direct killing assays, and incubated with shaking at
numerous Yp strains, Heine et al. [41], observed similar MIC results at 200 rpm and at the appropriate temperature. Samples were collected
both temperatures; most of the MICs were within one well at the two from the cultures at three to four time points, just prior to incubation
temperatures, with only 17.77% (8/45) of the 90% MICs having a two- (t0) and after two or three periods of incubation between 6 h and 72 h
well difference [41]. The MIC plates were read 18–24 h post incubation depending on the strain. Viable counts were determined by serial di-
(except 48 h for Yp and 72 h for Ft) and the MIC determined. The MIC lution and plating.
value was the lowest concentration of antimicrobial agent producing no
visible growth after incubation or the greatest reduction in growth 2.6. Statistics
density, as described below. Minimum bactericidal concentration
(MBC) values were ascertained by plating a 10% volume of the MIC The statistical significance (P < 0.05) of viable counts acquired in
assay wells lacking visible growth to determine the lowest anti- Direct Killing or Time Kill assays were determined by t-tests with the
microbial concentration producing no growth. Holm-Šídák method to correct for multiple comparisons, or by ANOVA
and pairwise multi-comparison post-test (e.g., Tukey's). These statistical
2.3.2. Checkerboard assay analyses were done with GraphPad Prism versions 7.0 or 8.1.
To identify combinations of an antibiotic and AMP which exhibited
synergistic or enhanced antibacterial activity, checkerboard MIC titra- 3. Results
tion assays were performed by a standard method [44–46]. Briefly,
each antimicrobial was serially titrated two-fold across all the rows or 3.1. Antimicrobial susceptibility to antibiotics and AMPs
down the columns in broth microdilution plates; the wells were com-
bined in a third tray which was inoculated with the test organism to MIC assays were performed to screen for antibiotics and AMPs

3
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

Table 2 Table 2 (continued)


Microbial sensitivity to antibiotics and antimicrobial peptides.
Bacterial strain Antimicrobial agent MICa - mean (range)b MBCa
Bacterial strain Antimicrobial agent MICa - mean (range)b MBCa

Antibiotics Peptides
Bp JW270 ceftazadime 3.91 Ndc Bp JW270 WLBU2 >62.5 (31.25->125) >62.5
E. coli ATCC 25922 0.092 (0.061–0.122) Nd E. coli ATCC 25922 5.60 (1.95–17.7) Ndc
Ba Sterne >31.2d >31.2 Ba Sterne 7.81 (3.91–15.62) Nd
Yp CO92 pgm- pPst- 0.122 0.122 Yp CO92 pgm- pPst- 31.25 31.25
Ft LVS strR 0.0305 Nd Ft LVS strR 9.76 (3.91–15.62) 31.25
Bm CLH001 3.91 (1.95–7.81) Nd Bm CLH001 62.5 (31.25->62.5) Nd

Bp JW270 fosfomycin > 62.5 Nd Bp JW270 BMAP18 >62.5 Nd


E. coli ATCC 25922 15.62 Nd E. coli ATCC 25922 7.81 Nd
Ba Sterne 31.25 Nd Ba Sterne 15.62 Nd
Yp CO92 pgm- pPst- 31.25 Nd Yp CO92 pgm- pPst >62.5 >62.5
Ft LVS strR > 62.5 Nd Ft LVS strR 1.95 Nd
Bm CLH001 > 62.5 Nd Bm CLH001 >62.5 Nd

Bp JW270 colistin > 62.5 Nd Bp JW270 Mastoparan7 >62.5 >62.5


E. coli ATCC 25922 1.95 Nd E. coli ATCC 25922 3.91 Nd
Ba Sterne 62.5 Nd
Yp CO92 pgm- pPst- >62.5 >62.5
Bp JW270 aztreonam 31.25 Nd
Ba Sterne >62.5 Nd
Yp CO92 pgm- pPst- 0.03 Nd Bp JW270 Nisin >625 (625–774) Nd
Ft LVS strR 31.25 Nd E. coli ATCC 25922 >625 (625–774) Nd
Bm CLH001 31.25 Nd

Bp JW270 Melittin >62.5 Nd


Bp JW270 nalidixic acid 7.81 (3.91–15.62) Nd E. coli ATCC 25922 62.5 Nd
E. coli ATCC 25922 1.95 (<0.98–1.95) Nd
Ba Sterne 3.91 Nd
Yp CO92 pgm- pPst- 15.62 Nd Bp JW270 Bactenicin >62.5 Nd
Ft LVS strR 0.487 Nd E. coli ATCC 25922 >62.5 Nd
Bm CLH001 31.25 Nd

Bp JW270 MagaininII >62.5 Nd


Bp JW270 minocycline 2.94 (1.95–3.91) >62.5 E. coli ATCC 25922 >62.5 Nd
E. coli ATCC 25922 0.102 (0.061–0.122) Nd
Ba Sterne 0.041 (0.031–0.98) >62.5
Yp CO92 pgm- pPst- 1.63 (0.98–1.95) >62.5 Bp JW270 CAMA >62.5 Nd
Ft LVS strR 0.061 Nd E. coli ATCC 25922 62.5 Nd
Bm CLH001 1.37 (0.98–1.95) Nd
a
The antimicrobial measures (MIC and MBC) are described in the Materials
and Methods. Values are μg/ml.
Bp JW270 doxycycline 2.93 (1.95–3.91) >31.25 b
Values with no range indicate identical MIC values were obtained in dif-
E. coli ATCC 25922 0.244 Nd
Ba Sterne 0.98 >62.5
ferent experiments.
c
Yp CO92 pgm- pPst- 0.98 >62.5 Nd - Not done.
d
Ft LVS strR 0.122 Nd The signs indicate limits of detection (LOD): ">" indicates the highest
Bm CLH001 0.98 Nd concentration tested, and "<" gives the lowest tested.

Bp JW270 tigecycline 2.93 (1.95–7.82) Nd


having potential use in broad-spectrum therapeutic combinations. Eight
E. coli ATCC 25922 0.244 Nd early generation antibiotics were selected, and MICs for surrogate
Ba Sterne 0.349 (0.12–0.98) >15.62 strains of Ba, Yp, Bp, Bm, and Ft (Table 1) were determined. Two ad-
Yp CO92 pgm- pPst- 0.98 (0.49–1.95) >15.62 ditional antibiotics in wide current use, doxycycline and ceftazidime,
Ft LVS strR 0.244 0.98
were included as controls. Three of the antibiotics had low to moderate
Bm CLH001 15.62 (7.81–31.25) Nd
MICs for the five strains and the Ec standard strain: novobiocin, tige-
cycline and minocycline; the latter two belong to the tetracycline fa-
Bp JW270 novobiocin 1.63 (0.98–1.95) Nd mily. Exceptions included the relative resistances of Bm to tigecycline
Ba Sterne 0.82 (0.49–1.95) Nd
(15.62 μg/ml) and Yp to novobiocin (31.25 μg/ml), as shown in
Yp CO92 pgm- pPst- 31.25 Nd
Ft LVS strR 0.046 Nd Table 2. The Burkholderia, were overall the most resistant of the five
Bm CLH001 1.30 (0.98–1.95) Nd species, as shown in Table 2. Most antibiotics had similar MICs for Bm
and Bp, however Bm was more resistant than Bp to nalidixic acid and
tigecycline, whereas Bp was more resistant to doxycycline and possibly
Bp JW270 fusidic acid 31.25 Nd
Bm CLH001 31.25 Nd
minocycline.
Of the eight peptides tested, only WLBU2 and BMAP-18 showed
activity against the Burkholderia and the other surrogate strains tested
(Table 2). Subsequent work centered on WLBU2, a cationic 24 amino
acid helical peptide which had been shown previously to exhibit broad-
spectrum activity against Ft, Bp, and Yp, and to be effective against
intracellular infection without host cell cytotoxicity [18,20]. It was

4
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

observed that in the MIC assays, AMPs such as WLBU2 and some bac- concentrations of the individual constituents thereby lessening their
teriostatic antibiotics did not completely eliminate production of visible toxicity. The checkerboard MIC data were analyzed by determining the
growth by the bacteria, especially the Burkholderia spp., but produced FICI values. The FICI results for the three combinations for five species
large reductions in growth. The MIC was defined as the concentration are available as supplementary tables (Tables A1, A.2, and A.3). Each of
which inhibited visible growth or produced the largest reduction in the three antibiotic-AMP combinations was effective (synergistic or ad-
growth density by visual inspection and with selected confirmation by ditive) against at least three strains, as determined by a reduction in MIC.
viable count determinations or OD600 readings [40–42]. Considered together, the minocycline- or tigecycline-peptide pairs in-
creased the antibacterial sensitivities of Yp, Ft, Ba and Bp (Table 3). The
3.2. Antimicrobial activity of therapeutic combinations novobiocin-WLBU2 combination augmented the susceptibility of all five
strains (Table 3). The Burkholderia were the most resistant strains overall,
3.2.1. Checkerboard MIC assays especially Bm; i.e., the two tetracycline-peptide combinations failed to
Based on these results, a preliminary set of effective candidates for increase Bm sensitivity beyond that of either alone. However, the high
evaluation as combination therapeutics was identified, which included sensitivity of Bm CLH001 to minocycline (and novobiocin) may have
the antibiotics tigecycline, minocycline, or novobiocin and peptide partially prevented any additional antibacterial effects of the peptide. In
WLBU2. The extent of synergistic or enhanced antibacterial activity of addition to the selected antimicrobial combinations, assays were per-
these down-selected combinations was characterized by three methods, formed to consider other potential antimicrobial candidates. Combina-
checkerboard MIC, direct killing assays, and time kill kinetic assays. tions of WLBU2 and nalidixic acid caused no change in the MIC for Ec but
Improved activity could potentially allow reductions in the effective afforded an 8-fold reduction in MIC for Bp; and the AMP nisin combined

Table 3
Summary of the effects on antibacterial activity of AMP-antibiotic combinations.

5
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

(Fig. 1). A WLBU2 dose of 0.25x MIC reduced Ba viability by >50%. The
Burkholderia were again the least sensitive of the five strains tested, as
predicted by the sensitivity results (Table 2). In agreement with previous
findings [18], Bp was partially killed by WLBU2, which caused ap-
proximately 35% killing at concentrations from 7.81 to 62.5 μg/ml. The
extent of direct killing of Bm by WLBU2 was also low, ranging from none
to a 37% reduction at high doses of ≥31.25 μg/ml.
To determine if the addition of an antibiotic could enhance direct
killing, the assays were performed with the peptide in combination with
tigecycline, minocycline, or novobiocin. Antibiotics with bacteriostatic
activity such as the tetracyclines are often only effective against re-
plicating and/or metabolizing bacteria [30,51]. However, tetracycline
can alter cytosolic membranes and directly cause cell leakage [52], and
DNA gyrase inhibitors such as novobiocin can exhibit concentration-de-
pendent bactericidal effects [53]. Furthermore, the effects of WLBU2 on
membrane integrity could potentially increase antibiotic uptake [4,54].
The results of the direct killing assays partially supported the enhanced
antibacterial efficacy of the antibiotic-peptide combinations (combos)
and emphasized the importance of identifying the optimal concentrations
Fig. 1. Direct killing of Ba by peptide WLBU2 in buffer. Germinated spores of of both components. As summarized in Table 3, the direct killing data did
Ba were suspended in PBS, incubated with dilutions of the AMP, and plated for not always parallel the MIC data (using growing cultures), as expected.
viable counts. Data are the mean CFU/ml and SD (error bars). All five con- The tetracycline-peptide combos augmented direct killing of Ba, Bp, and
centrations of WLBU2 reduced Ba viable counts compared to the untreated Bm; and the novobiocin-WLBU2 combo enhanced killing by Bp. None of
control, p < 0.0001. From highest to lowest WLBU2 concentrations, viable the combos augmented the direct killing of Yp, which was very sensitive
counts ranged from 17.6% to 50.1% of the diluent control. The difference be-
to WLBU2 alone in the three assays (Table 3). Fig. 2 illustrates the in-
tween mean counts of the highest (31.25 μg/ml WLBU2) and lowest (1.95 μg/
crease in killing of Bp and Bm afforded by combining tigecycline and
ml) dose groups was significant, p = 0.0017.
WLBU2 in the checkerboard and killing assays. Whereas tigecycline
alone significantly reduced Bp viability (p < 0.0001), WLBU2 did not
with either colistin or nalidixic acid resulted in no change in antibacterial (Fig. 2A). The tigecycline-associated killing was further enhanced by
activity for Ec or Bp (data not shown). combining 0.98 μg/ml tigecycline (0.33x MIC) with WLBU2
(p = 0.0009) but not by the other four combo treatments. Thus, effects
3.2.2. Direct killing tests of the peptide were dependent on concentration but was not directly
Antimicrobial peptides have been shown to directly kill bacteria in dose-related. The results suggested that the activity for Bp of a low
the absence of growth [1,4,5,8,18,20]. To assess direct killing by WLBU2, concentration of tigecycline (0.33x MIC) could be significantly enhanced
fresh cultures of the bacteria were suspended in buffer (2.4), incubated by the addition of WLBU2. Also, the reduced bactericidal activity of the
for 2 h with different peptide concentrations, and dilution plated for combos containing higher concentrations of tigecycline (p = 0.002 for
viable counts. The bactericidal activity of the peptide was not always the 3.91 μg/ml combo) suggested that the peptide might have antag-
proportional to the dose and inter-experimental variability was occa- onistic effects on tigecycline depending on the concentration of each.
sionally observed. However, the inclusion of untreated controls in all Whereas neither antimicrobial alone significantly impacted Bm viability
experiments allowed the effects of the treatments on viability to be de- (Fig. 2B), the peptide combined with tigecycline from 3.91 to 15.62 μg/
termined within each experiment. As reported previously [18], our ml significantly increased killing.
strains of Yp and Ft were sensitive to killing by WLBU2. Treatment of Ft
with 15.62 μg/ml WLBU2 caused a mean of 61% killing compared to the 3.2.3. Time kill assays
control. Exposure of Yp to 0.98 μg/ml (0.03x MIC) resulted in a 70% To analyze time-associated antimicrobial killing, flasks with MHB,
reduction in viability in these tests compared to the untreated control alone or with antimicrobials present, were inoculated and cultures were
(data not shown). Similar to Yp, Ba was also very sensitive to the peptide sampled at three to four time points for viable counts. The findings are

Fig. 2. Direct killing of Burkholderia by tigecycline


and peptide WLBU2. A. Bp. WLBU2 and tigecy-
cline were tested separately (31.25 μg/ml or
0.98 μg/ml, respectively), or in five combinations.
The combos contained WLBU2 (31.25 μg/ml) and
tigecycline ranging from 0.015 μg/ml to 3.91 μg/
ml, as indicated in the X axes. The combo reduced
viability more than did tigecycline alone
(*p = 0.009). B. Bm. The combos contained
WLBU2 (31.25 μg/ml). WLBU2 alone and tigecy-
cline alone (3.91 μg/ml or 15.62 μg/ml) did not
significantly reduce the viable counts compared to
the control. The combos with tigecycline from
3.91 to 15.62 μg/ml significantly reduced viability
compared to the control (**p ≤ 0.002).

6
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

Fig. 3. Time course assays of killing of three strains by a tetracycline-peptide combination. The change in viable counts with incubation time in MHB with or without
an antibiotic or WLBU2 are shown. A. Yp pgm-pPst-was treated with minocycline (1.46 μg/ml) and/or WLBU2 (3.91 μg/ml). The combo group counts were less than
the other groups at 24 h and 48 h, with p values from 0.0087 to <0.0001. B. Ft LVS was treated with tigecycline (0.488 μg/ml) and WLBU2 (3.78 μg/ml). The combo
treatment counts were less than those of the other groups for the 24 h and 72 h samples, p values from <0.0001 to 0.033. C. Ba was treated with minocycline
(0.328 μg/ml) and/or WLBU2 (1.96 μg/ml, 0.25x MIC). The counts of the combo-treated group were less or nearly significantly less than those of minocycline alone
at 6 h and 24 h (p < 0.0001) and WLBU2 alone at 6 h (p = 0.063) and 24 h (p < 0.0001).

Fig. 4. Time course assays of killing of Bp by novobiocin-peptide combinations: effects of growth conditions on antibacterial activity. A. Treatment with WLBU2 and
novobiocin (6.52 μg/ml) and incubation in standard conditions. The viable counts of the combo at 24 h were less than those of the novobiocin alone (p = 0.0076)
and the WLBU2 or untreated groups (p = 0.0001). B and C. Treatment with WLBU2 and novobiocin (13.04 μg/ml). B. Incubation was in MHB prepared by standard
procedures. WLBU2 treatment had no effect on killing. C. Incubation was in nutritionally suboptimal MHB. At 24 h and 48 h, the combo counts were reduced
compared to those of the WLBU2 treated (p ≤ 0.009) and of the untreated and novobiocin treated groups (p ≤ 0.0058).

summarized in Table 3 and illustrated in Fig. 3, Fig. 4, and Fig. 5. The treatments were bactericidal for Yp at 24 h, and the combo continued
tetracycline-AMP combos had significantly enhanced antibacterial ac- killing to 48 h (Fig. 3A). WLBU2 alone temporarily retarded growth to
tivity against Ba, Ft, and Yp (Fig. 3). The minocycline alone and combo 24 h. The tigecycline-WLBU2 combination significantly impeded

7
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

Fig. 5. Time course assays of killing of Bm by antibiotic-peptide combinations. The change in viable counts of Bm with incubation time in FeSO4-supplemented MHB.
A. WLBU2 (31.25 μg/ml) had no effect on, or antagonized killing by, novobiocin (6.53 μg/ml) after 48 h incubation. B. Treatment with tigecycline (15.62 μg/ml) and
WLBU2 (7.81 μg/ml). At 6 h, the combo counts were less than those of the WLBU2-treated and untreated (p = 0.0004) but not the tigecycline-treated (p = 0.241)
samples. At 24 h, the combo counts were less than those of tigecycline (p = 0.0021), WLBU2 (p < 0.0001), and untreated (p = 0.0004) samples. C. The conditions
were the same as B except the iron concentration was decreased two-fold. Tigecycline killing was increased 240–fold compared to that in panel B.

growth of Ft LVS compared to the other three groups at 24 h and 72 h antibacterial effects of the therapeutics observed in vitro might differ
(Fig. 3B). A similar enhancing effect of a minocycline-WLBU2 combo from those occurring in the less optimal growth environment.
was observed with Ba (Fig. 3C). Compared to the other three groups,
the combo treatment caused the greatest reduction in viability at both 4. Discussion
the early (6 h) and last timepoint (Fig. 3C). The combo-treated values
remained less than those of the minocycline-treated, though the two- In this study, antibiotic-AMP combinations were identified as po-
fold difference was no longer significant by 24 h. tentially useful alternative countermeasures for treating infections by
Neither tetracycline-peptide combo positively impacted Bp killing, five bacterial biothreat species, including the AMR Burkholderia bac-
and in some circumstances antagonized the antibacterial effects of the teria. The three down-selected combinations consisted of a tetracycline
antibiotic (Table 3). However, the novobiocin-WLBU2 combo sig- (tigecycline or minocycline) or gyrase inhibitor novobiocin, each
nificantly reduced viability of all five strains except Bm (Table 3). As combined with a cationic AMP (WLBU2). No single antibiotic-AMP
shown in Fig. 4 (and Table 3), peptide WLBU2 enhanced the killing of combination exhibited synergistic or enhanced antibacterial activity
Bp in combination with a lower concentration of novobiocin (Fig. 4A) against all five bacterial agents in all three assays. However, based on
or had no effect on killing associated with a larger concentration the results of these antimicrobial sensitivity measures, at least one
(Fig. 4B). The performance of the assay under suboptimal growth specific treatment (and an alternate) were effective for each strain: AMP
conditions greatly enhanced the killing of Bp by novobiocin-peptide. In with novobiocin (or with minocycline) for Ba and Yp, AMP with no-
Fig. 4C, Bp was incubated in MHB medium which had been subjected to vobiocin (or tigecycline alone) for Bp, AMP with novobiocin (or with
a longer than recommended period of steam sterilization. It was nu- tigecycline) for Ft, and AMP with tigecycline for Bm. Novobiocin alone
tritionally inadequate as shown by the absence of growth by the un- was also an effective alternate antimicrobial for Bm, but due to toxicity
treated control after 6 h. However, at 24 h and 48 h, the combo counts associated with this antibiotic (discussed below), this is not a realistic
were greatly reduced compared to the other three groups. The growth therapeutic option.
conditions described in Fig. 4C could resemble that occurring in in vivo The advantages of managing complex infections with a combination
niches which induce a dormant or persistent bacterial state such as the of antibiotics is well established [55]. This strategy is valuable when the
host cell phagolysosome. In such circumstances, combination anti- components display synergistic or additive effects in vitro; the microbial
microbials might be especially effective therapeutics, as elaborated etiology cannot be determined, is polymicrobial, or is caused by an
upon later. Fig. 5 illustrates the effects of two antibiotic-peptide combos AMR strain; or the infection is prone to an extended course and emer-
on Bm viability. Bm CLH001 was very sensitive to novobiocin (Fig. 5A). gence of resistance. The efficacy of antibiotic combination therapy is
WLBU2 did not augment killing but instead appeared to antagonize supported by studies performed in clinical and in vivo settings
antibiotic killing at high concentration. In contrast, the peptide en- [48,49,55–58]. However, the current escalation in bacterial resistance
hanced the tigecycline-associated decrease in Bm viability (Fig. 5B). to conventional antibiotics has stimulated an urgent search for alternate
Interestingly, when the iron-deficient Bm CLH001 was incubated in therapeutics.
medium with a suboptimal iron concentration, the peptide did not Antimicrobial peptides are good candidates to consider in an al-
enhance tigecycline killing, however, the activity of tigecycline alone ternate antibiotic-AMP dual treatment approach. Hallmarks of AMPs
was increased 240–fold (Fig. 5C). These findings again suggested that include their broad and rapid activity against a diverse range of

8
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

pathogens. The mechanisms, which can be both direct and indirect, substituted (e.g., fluorinated) amino acids, and the modification of
underlying the potent antimicrobial action of AMPs have been widely peptides by polymers [9,14,19,20,62,63,70,87,88]. Also, AMPs can be
studied [1,2,59]. The major mode of direct bacterial cytotoxicity occurs rendered less harmful to the host by minimizing their interaction with
by perturbation of the bacterial outer membrane. Natural AMPs com- host tissues. Epithelial cells are more resistant to AMP toxicity and in-
monly have an amphipathic conformation with positively charged and jection of the peptides intracutaneously might obviate some of their
hydrophobic groups present on opposing faces of the structure, which toxicity [87]. Similarly, encapsulation and delivery of AMPs to pha-
imparts the ability to electrostatically disrupt a phospholipid membrane gocytic leukocytes could conceivably allow a peptide to primarily bind
as found on bacterial surfaces [1,2,10]. The mechanisms for this are not to and kill phagocytosed bacteria. A peptide optimization approach is
well understood but models describe the creation of membrane pores discussed further below for WLBU2.
with subsequent insertion of the AMP and leakage of ions and meta- Finally, since AMPs are often insoluble in water and must be dis-
bolites and ultimately the loss of membrane function; alternately, solved in a solvent such as DMSO, it is important to consider the anti-
membrane absorption of the AMPs as a layer, followed by membrane host effects of residual levels of solvent in the antimicrobial inocula. In
solubilization and disintegration, has been described [1–3,59]. Some discerning the most effective combinations of AMP and antibiotic in our
AMPs penetrate cells by other means which involve bacterial cell pe- study, we selected those with total final DMSO concentrations below
netration as a result of the affinity of AMPs for LPS or the facilitation by those which inhibited the bacteria, as indicated above. The toxicity of
proline moieties of phospholipid interactions by AMPs [2,60]. Upon cell DMSO for humans and animals is also a concern but is not well defined.
entry, the AMPS interfere with critical intracellular functions including It is considered to be slightly hazardous for humans in case of inhala-
RNA and protein synthesis and other processes [2,61]. These various tion and contact of full strength DMSO with the skin and eyes [89,90].
anti-bacterial activities of AMPs are not mutually exclusive, a property It has been approved for use by the FDA for specific clinical conditions
which likely affords AMPs the ability to evade bacterial resistance such as inflammatory genitourinary disorders and has been used to
mechanisms [62]. treat numerous other conditions [90,91]. In mice DMSO has a max-
In addition to the direct bacterial killing, many AMPs exhibit in- imum tolerated intravenous dose (MTD) of 40% (2200 mg/kg) and a
direct function through immunomodulatory activities, i.e., the stimu- maximum to-observed-effect dose (NOEL) of 30% (1650 mg/kg)
lation of chemotaxis, differentiation of immune cells, and regulation of [92,93]. In our combination assays, DMSO was used to prepare con-
inflammatory cell responses and cell death pathways. Related host- centrated stocks of WLBU2, and residual levels of the solvent were
protective functions include the ability to neutralize LPS and disrupt present in final amounts ranging from 0.04% to 0.625%, as discussed
biofilms [1,2,59,63]. Both the direct and indirect AMP actions can further below.
contribute to protective host responses to invading pathogens. Thus, The combination of AMPs with an antibiotic may serve to reduce the
AMPs are often referred to as host defense peptides (HDP). toxicity and off-target activity of the peptide. Naghmouchi and cow-
In contrast to conventional antibiotics, AMPs have a number of orkers showed that combinations of the peptides nisin A or pediocin
properties that confer the ability to overcome common mechanisms of with colistin were synergistic, thus permitting a reduction in the ef-
multi-antibiotic resistant pathogens. Many AMPs do not require meta- fective concentrations (and thus toxicities) of each component [23];
bolically active, replicating organisms to kill; and they inactivate bac- this finding was confirmed and extended in recent studies employing
teria rapidly which limits time for mutants to predominate. Also, some other AMPs and standard antibiotic drugs of choice [54].
AMPs act on more than one host target, as mentioned above Although synergistic combinations of an antibiotic and AMP pro-
[2,54,59,64–66]. However, bacteria can develop resistance to AMPs, vide a promising approach for combatting infections, the basis of this
though this is expected to be much less common than resistance to synergy is not well understood. A discussion of the many established
antibiotics and more difficult for bacteria to acquire [2,3,54,59]. As an and proposed mechanisms is beyond the scope of this paper and are
example of an AMP resistance mechanism, changes in bacterial outer reviewed in detail elsewhere [54]. The major way by which AMPs with
membrane lipoteichoic acid can occur which reduce its negative charge strong membrane permeabilizing activity enhance susceptibility is to
and thus binding by AMPs [3]. increase the access of antibiotics to their host cell targets; yet as noted
Although AMPs have long been attractive as potential anti- above, some AMPs can act solely or primarily on internal bacterial
microbials due to their aforementioned properties, there are few reports targets. Furthermore, the synergy observed for antibiotic-AMPs is not
demonstrating actual therapeutic in vivo efficacy of AMPs. Protection of necessarily unidirectional. In addition to the positive effects of AMPs on
mice was afforded by defensins against lethal infection with P. aerugi- antibiotic activity, the antibiotic may cause alterations in the host cell
nosa; infection by Ba spores or exposure to anthrax lethal toxin; and which, for instance, improve the incorporation or orientation of AMPs
infection by carbapenem-resistant clinical isolates of Acinetobacter in the bacterial membrane [54].
baumanni [4,19,64,67,68]. Although various AMPs are in human clin- In the current study, we distinguished licensed but rarely used an-
ical trials, only a few FDA-approved products are available, though tibiotics which in combination with an AMP enhanced the susceptibility
rarely used (e.g., the polymixins). This paucity of efficacy data is par- of five Tier 1 select agents. Three candidates were identified as proto-
tially due to toxicity concerns and also to other known and potential types of effective broad spectrum therapeutics, and consisted of the
drawbacks of the natural AMPs. For instance, some AMPs lack specifi- peptide WLBU2 and the antibiotics tigecycline, minocycline, or novo-
city, e.g., bind unintended host targets in tissues and serum, leading to biocin. The eight AMPs screened initially were chosen based on the
toxic effects as manifested, i.e., by lysis of erythrocytes due to mem- literature, as described in the Introduction and previous studies
brane permeation and disruption [16,54,87]. Furthermore, their anti- [1,5,7,18,70], with specific emphasis on Burkholderia spp. due to their
microbial activity can be inhibited in the presence of acidic pH, sodium high-level of intrinsic antibiotic resistance. In killing assays with ten
ions, divalent cations, proteases, and other substances, thus reducing AMPs, Kanthawong detected low to moderate sensitivity to killing of Bp
the level of free peptide available for antimicrobial use [16,17,69,87]. but at high concentrations only (100 μM–200 μM). The exception was
These limitations can be surmounted to a large extent by structural LL37, which caused a large, rapid decrease in viability of Bp exposed to
optimization. AMP modifications can be made which theoretically 200 μM peptide. This LL37 sensitivity was not observed by other
cause them to be antibacterial at concentrations well below those which workers and is probably Bp strain-related [5,7,18].
are lytic for host cells and also to more narrowly target bacteria The engineered peptide WLBU2 was down-selected as the AMP
[9,14,19,20,62,63,70,87,88]. Several approaches have also been uti- component of a combination therapeutic based on the MIC and
lized to improve the cell selectivity of AMPs. These include the opti- checkerboard assay findings. The engineering of novel peptides from
mization of physicochemical properties of peptides, constraint of AMP native progenitors to maximize potency and specificity for anti-
conformations, the use of unusual amino acids or of D-stereoisomer or microbial targets is an established tactic [9,14,19,20,70]. Unlike six of

9
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

the seven other AMPs, WLBU2 displayed antibacterial activity against has an extended range against gram-positive and –negative bacteria
all five agents (Table 2; and data described in the text). The suscept- [73]. Due to its large additional side chain, it can avoid some of the
ibilities of the five biothreat species were greater for WLBU2 than for common tetracycline resistance mechanisms and is active against many
BMAP-18, except for Ft LVS. Its selection is also supported by previous tetracycline-resistant strains. Specifically it can overcome resistance
work to construct and characterize WLBU2. This 24-aa cationic peptide, caused by at least one of the tetracycline efflux pumps and by the
composed of valine, arginine, and tryptophan residues, was one of bacterial ribosome protection protein, but not due to the tetracycline
several derivatives of a lytic peptide base. WLBU2 had the highest ac- inactivation enzyme or the RND antibiotic efflux pump [73,74]. It has
tivity against Pa. It possessed broad spectrum activity against Yp, Ft, improved the outcome of infection with carbapenem-resistant and XDR
and Bp and killed intracellular Ft in macrophage assays [18]. The Acinetobacter and Klebsiella, when used together with a second anti-
peptide retained its activity in serum and blood; killed Pa in co-cultures biotic [56–58]). The potential disadvantages of tigecycline include its
with human skin fibroblast without cell toxicity; prevented biofilms on bacteriostatic activity in vitro, the possibility of bacterial evolution to
primary human airway epithelial cells; and protected mice against in- resistance [74], and reported adverse events and clinical failures with
fection when administered by intravenous or aerosol routes treatment [75]. In the current study, the five strains were all very
[4,19,20,63]. The structure of and the rate of killing by WLBU2 sup- sensitive to tigecycline except for Bm (Tables 2 and 3). The strain of Bm
ported the conclusion that it activity was due primarily to direct used, Bm CLH001 ΔtonB, Δhcp1, is becoming widely used as a surrogate,
membrane disruption, as occurs for the human AMP LL37 [1,71]. however wild type Bm strains are reported to be sensitive to tigecycline,
However it is possible that the peptide has ancillary intracellular effects e.g., a panel of 23 human and equine clinical isolates of Bm had MICs
on bacterial processes (synthesis, transcription or translation), as de- ranging from 0.01 to 0.25 μg/ml (unpublished data, USAMRIID Ther-
scribed for some defensins and cathelicidins [61,72] or it has other apeutics core). It is conceivable that an alteration occurred in the outer
immunomodulatory effects on host responses. Studies have docu- membrane of the mutant strain which impeded uptake of the antibiotic.
mented both the loss and retention of WLBU2 activity in the presence of Therefore, tigecycline may be even more broadly efficacious as part of a
RBCs [16,19]. The importance of this or other potential limitations to combination therapeutic against wild type Bm than described here.
its clinical use may require further tests of in vivo efficacy in animal Minocycline is also broadly effective, has a good pharmacokinetic
models. profile and is typically safe and well-tolerated, although possibly less so
Based on previous in vitro and in vivo data, there is ample evidence than doxycycline [76]. However, it was among the most effective an-
to support the premise that clinical efficacy of the peptide-antibiotic tibiotics against three groups which are often difficult to treat, multiply
combinations against biothreat pathogens is possible at achievable yet AMR strains of the Acinetobacter spp complex, the Burkholderia cepacia
nontoxic doses. As described above, Deslouches and coworkers complex, and Stenotrophomonas maltophilia [77]. Also, minocycline has
[4,18–20,59,63,94,95] have evaluated the effects of WLBU2 on multi- intriguing non-antibiotic biological effects (anti-inflammatory, im-
drug resistant “ESKAPE” pathogens such as Pa, a common pulmonary munomodulatory, and neuroprotective) [76,78], a property which may
and systemic pathogen which is often refractory to treatment. In later augment its value in treating infected patients who are immuno-com-
studies, as little as 16–20 μM WLBU2 prevented Pa biofilm formation on promised [78]. In the current study, all five species were highly sus-
primary human airway epithelial cells; and concentrations up to at least ceptible to minocycline with MICs ranging from 0.06 to 2.94 μg/ml.
50 μM were not toxic for the cells as shown by no change in transe- Novobiocin is a member of the aminocoumarin class of antibiotics
pithelial electrical resistance [4,63]. Furthermore, WLBU2 (20 μM) also [79,80]. It is a bacterial DNA gyrase inhibitor and acts by inhibiting the
significantly enhanced killing of Pa in this biofilm model by sub- GyrB subunit of the enzyme. Novobiocin is bacteriostatic and may be
inhibitory concentrations of antibiotics which are currently used in bactericidal at higher concentrations; however it is rarely used clini-
clinical treatment of biothreat agents (ciprofloxacin, meropenem, cef- cally, due to reasons of safety and efficacy, and diminishing commercial
tazidime). Finally, Chen et al. achieved protection of mice against antibiotic development efforts [79]. The potency of novobiocin is
pneumonia by WLBU2 delivered intratracheally after infection with Pa considerably higher than that of the fluoroquinolones which also target
[4]. The lowest therapeutic dose was determined to be equivalent to DNA gyrase, but at a different site on the enzyme. Novobiocin is active
0.05 mg/kg (1.25 μg/25 gm mouse), which was 280-fold below the mostly against gram-positive bacteria and is an effective anti-staphy-
maximum tolerated (nontoxic) dose of peptide, determined to be lococcal agent used to treat MRSA, but it has reduced activity against
14 mg/kg [94] in mice given WLBU2 intraperitoneally or intravenously many gram-negatives due to their LPS-associated membrane perme-
after exposure to a lethal dose of Pa. WLBU2 reduced both the bacterial ability barrier or to specific resistance mechanisms [31,79]. Never-
load (by 2 logs) and the extent of its induced inflammation with no theless, we observed low novobiocin MICs for not only Ba but also for
signs of disease or AMP-associated toxicity. Recently, Abdelbaqi and Bp, Bm, and Ft (Table 2), in agreement with previous reports [42,81]. In
coworkers demonstrated efficacy of the peptide against Bp, Ft, and Yp; contrast, Yp was novobiocin-resistant, as described by Heine et al. [41].
and the killing of Ft within infected macrophages with WLBU2 was Interestingly, in a screen with a small molecule library, Taylor and
achieved by 12.5 μM–25 μM [18]. In our assays, antibiotic activity coworkers found compounds, termed “antibiotic adjuvants”, which
against the five bacterial agents was significantly enhanced by WLBU2 when combined with novobiocin, appeared to permeabilize the bac-
at 2.0–31.25 μg/ml (approximately 0.5–10 μM), which is well within terial membrane, facilitating novobiocin entry and lowering the MIC
the range of effective concentrations of this peptide as described above. [30]. Similarly, Savage [31] described the ability of cationic outer
The three antibiotics selected for evaluation as the antibiotic com- membrane permeabilizers to sensitize gram-negative bacteria to hy-
ponent of combination therapeutics were chosen based on their prop- drophobic antibiotics such as novobiocin and thus extend the anti-
erties and their activity in this study. They are older licensed antibiotics bacterial spectrum of these antibiotics [30,31,79]. The enhanced anti-
which are not in high use currently, but have the potential for inclusion microbial activity of novobiocin-AMP combinations against the
in combination therapies. The results of MIC screening, checkerboard pathogens, to include Yp resembles that of these antibiotic-enhancing
assays, and functional tests supported the latter premise. substances.
Tigecycline and minocycline are members of the tetracycline family, Infections which are complicated and difficult to treat with con-
broad spectrum polyketide antibiotics produced by Streptomyces which ventional antibiotics include those caused by bacteria which can sur-
are bacteriostatic protein synthesis inhibitors. They inhibit the initia- vive within host cells or other in vivo niche in a non-growing but viable
tion of translation in various ways by attaching reversibly to the 30S state [51]. During certain stages of infection, the five bacterial agents
ribosomal subunit (composed of 16S rRNA and 21 proteins) and then exist in host cells such as macrophages, and in other in vivo niches such
inhibiting the binding of aminoacyl-tRNA to the mRNA translation as biofilms, in a non-replicating but persistent state [82–86]. Bacteria in
complex [43,49,53,55]. Tigecycline is a glycylcycline derivative which these settings are often antibiotic resistant. AMPs which have direct

10
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

killing activity may be useful therapeutic components for viable but [7] R.C. Bernhards, et al., Characterization of in vitro phenotypes of Burkholderia
dormant bacteria, and their lethal effects might be enhanced by con- pseudomallei and Burkholderia mallei strains potentially associated with persistent
infection in mice, Arch. Microbiol. 199 (2) (2017) 277–301.
comitant treatment with some antibiotics as suggested (Table 3 and [8] S. Tandhavanant, et al., Effect of colony morphology variation of Burkholderia
Fig. 4C). This scenario is supported by pharmacodynamic character- pseudomallei on intracellular survival and resistance to antimicrobial environments
istics of antibiotics which can cause prolonged antibacterial effects in in human macrophages in vitro, BMC Microbiol. 10 (2010) 303.
[9] M.A. Fox, et al., Design and characterization of novel hybrid antimicrobial peptides
vivo on slow- or non-growing bacteria. When serum levels of a bacter- based on cecropin A, LL-37 and magainin II, Peptides 33 (2) (2012) 197–205.
iostatic antibiotic fall below the MIC, antibacterial effects may linger [10] D. Andreu, L. Rivas, Animal antimicrobial peptides: an overview, Biopolymers 47
due to: persistent suppression of bacterial growth by the therapeutic, (6) (1998) 415–433.
[11] T. Higashijima, et al., Mastoparan, a peptide toxin from wasp venom, mimics re-
the enhanced susceptibility of treated bacteria to phagocyte killing, ceptors by activating GTP-binding regulatory proteins (G proteins), J. Biol. Chem.
and/or the ability of sub-MIC concentrations of antibiotics to alter 263 (14) (1988) 6491–6494.
bacterial morphology, slow the bacterial growth rate and extend anti- [12] B. Skerlavaj, et al., Biological characterization of two novel cathelicidin-derived
peptides and identification of structural requirements for their antimicrobial and
biotic suppression [43].
cell lytic activities, J. Biol. Chem. 271 (45) (1996) 28375–28381.
In conclusion, we identified candidate multi-spectrum therapeutics [13] X. Vila-Farres, et al., In vitro activity of several antimicrobial peptides against co-
consisting of specific combinations of an underutilized antibiotic and listin-susceptible and colistin-resistant Acinetobacter baumannii, Clin. Microbiol.
antimicrobial peptide which were active against a diverse set of bio- Infect. 18 (4) (2012) 383–387.
[14] M. Wu, R.E. Hancock, Improved derivatives of bactenecin, a cyclic dodecameric
threat agents. Subsequent efforts will utilize fully virulent strains of the antimicrobial cationic peptide, Antimicrob. Agents Chemother. 43 (5) (1999)
agents to investigate protective efficacy, host cytotoxicity, and in vivo 1274–1276.
availability of the candidates in cell culture and animal models [15] K. Madhongsa, et al., Antimicrobial action of the cyclic peptide bactenecin on
Burkholderia pseudomallei correlates with efficient membrane permeabilization,
[18,19,63,87,88]. Finally, it is conceivable that a triple antimicrobial PLoS Neglected Trop. Dis. 7 (6) (2013) e2267.
cocktail of the AMP, novobiocin, and either tigecycline or minocycline [16] C.G. Starr, J. He, W.C. Wimley, Host cell interactions are a significant barrier to the
might increase synergy due to the actions of a protein synthesis in- clinical utility of peptide antibiotics, ACS Chem. Biol. 11 (12) (2016) 3391–3399.
[17] C.G. Starr, W.C. Wimley, Antimicrobial peptides are degraded by the cytosolic
hibitor and a DNA gyrase inhibitor in combination with the AMP. proteases of human erythrocytes, Biochim. Biophys. Acta Biomembr. 1859 (12)
(2017) 2319–2326.
CRediT authorship contribution statement [18] S. Abdelbaqi, et al., Novel engineered cationic antimicrobial peptides display broad-
spectrum activity against Francisella tularensis, Yersinia pestis and Burkholderia
pseudomallei, J. Med. Microbiol. 65 (2) (2016) 188–194.
Christopher K. Cote: Investigation, Writing - review & editing. [19] B. Deslouches, et al., Activity of the de novo engineered antimicrobial peptide
Irma I. Blanco: Investigation. Melissa Hunter: Investigation. Jennifer WLBU2 against Pseudomonas aeruginosa in human serum and whole blood: im-
plications for systemic applications, Antimicrob. Agents Chemother. 49 (8) (2005)
L. Shoe: Investigation. Christopher P. Klimko: Investigation. Rekha
3208–3216.
G. Panchal: Supervision, Methodology, Funding acquisition. Susan L. [20] B. Deslouches, et al., De novo generation of cationic antimicrobial peptides: influ-
Welkos: Investigation, Conceptualization, Methodology, Data curation, ence of length and tryptophan substitution on antimicrobial activity, Antimicrob.
Writing - review & editing, Writing - original draft, Visualization, Agents Chemother. 49 (1) (2005) 316–322.
[21] D. Drider, S. Rebuffat, Prokaryotic Antimicrobial Peptides: from Genes to
Supervision, Funding acquisition, Formal analysis, Project administra- Applications, Springer, New York, NY, 2011.
tion, Resources. [22] B.P. Goldstein, et al., Activity of nisin against Streptococcus pneumoniae, in vitro,
and in a mouse infection model, J. Antimicrob. Chemother. 42 (2) (1998) 277–278.
[23] K. Naghmouchi, et al., Synergistic effect between colistin and bacteriocins in con-
Declaration of competing interest trolling Gram-negative pathogens and their potential to reduce antibiotic toxicity in
mammalian epithelial cells, Antimicrob. Agents Chemother. 57 (6) (2013)
The authors have no conflicts of interest to disclose. 2719–2725.
[24] Centers for Disease Control and Prevention DHHS, Possession, use, and transfer of
select agents and toxins; biennial review. Final rule, Fed. Regist. 7 (12) (2012)
Acknowledgments 1389.
[25] J. Garau, Other antimicrobials of interest in the era of extended-spectrum beta-
lactamases: fosfomycin, nitrofurantoin and tigecycline, Clin. Microbiol. Infect. 14
The authors thank Drs. J. Bozue and S. Biryukov for their critical
(Suppl 1) (2008) 198–202.
reading of this manuscript. [26] M.E. Falagas, A.P. Grammatikos, A. Michalopoulos, Potential of old-generation
Funding was provided by JSTO-CBD/DTRA Project CB10647- antibiotics to address current need for new antibiotics, Expert Rev. Anti Infect.
Ther. 6 (5) (2008) 593–600.
Bacterial Biothreat Therapeutics. Opinions, interpretations, conclu-
[27] Y. Kobayashi, H. Uchida, Y. Kawakami, Synergy with aztreonam and arbekacin or
sions, and recommendations are those of the authors and are not ne- tobramycin against Pseudomonas aeruginosa isolated from blood, J. Antimicrob.
cessarily endorsed by the U.S. Army. Chemother. 30 (6) (1992) 871–872.
[28] M. Popovic, et al., Fosfomycin: an old, new friend? Eur. J. Clin. Microbiol. Infect.
Dis. 29 (2) (2010) 127–142.
Appendix A. Supplementary data [29] K. Christiansen, Fusidic acid adverse drug reactions, Int. J. Antimicrob. Agents 12
(Suppl 2) (1999) S3–S9.
Supplementary data to this article can be found online at https:// [30] P.L. Taylor, et al., A forward chemical screen identifies antibiotic adjuvants in
Escherichia coli, ACS Chem. Biol. 7 (9) (2012) 1547–1555.
doi.org/10.1016/j.micpath.2020.104050. [31] P.B. Savage, Multidrug-resistant bacteria: overcoming antibiotic permeability bar-
riers of gram-negative bacteria, Ann. Med. 33 (3) (2001) 167–171.
References [32] T.M. Mott, et al., Characterization of the Burkholderia mallei tonB mutant and its
potential as a backbone strain for vaccine development, PLoS Neglected Trop. Dis. 9
(6) (2015) e0003863.
[1] F. Findlay, et al., Cationic host defense peptides; novel antimicrobial therapeutics [33] M. Sterne, Distribution and economic importance of anthrax, Fed. Proc. 26 (5)
against Category A pathogens and emerging infections, Pathog. Glob. Health 110 (1967) 1493–1495.
(4–5) (2016) 137–147. [34] J.M. Warawa, et al., Role for the Burkholderia pseudomallei capsular poly-
[2] M. Mahlapuu, et al., Antimicrobial peptides: an emerging category of therapeutic saccharide encoded by the wcb operon in acute disseminated melioidosis, Infect.
agents, Front. Cell Infect. Microbiol. 6 (2016) 194. Immun. 77 (12) (2009) 5252–5261.
[3] D.I. Andersson, D. Hughes, J.Z. Kubicek-Sutherland, Mechanisms and consequences [35] S. Saslaw, et al., Tularemia vaccine study. II. Respiratory challenge, Arch. Intern.
of bacterial resistance to antimicrobial peptides, Drug Resist. Updates 26 (2016) Med. 107 (1961) 702–714.
43–57. [36] S.L. Welkos, A.M. Friedlander, K.J. Davis, Studies on the role of plasminogen ac-
[4] C. Chen, et al., Enhanced efficacy of the engineered antimicrobial peptide WLBU2 tivator in systemic infection by virulent Yersinia pestis strain C092, Microb. Pathog.
via direct airway delivery in a murine model of Pseudomonas aeruginosa pneu- 23 (4) (1997) 211–223.
monia, Clin. Microbiol. Infect. 24 (5) (2018) 547 e1–547 e8. [37] S. Welkos, et al., Determination of the virulence of the pigmentation-deficient and
[5] S. Kanthawong, et al., In vitro susceptibility of Burkholderia pseudomallei to an- pigmentation-/plasminogen activator-deficient strains of Yersinia pestis in non-
timicrobial peptides, Int. J. Antimicrob. Agents 34 (4) (2009) 309–314. human primate and mouse models of pneumonic plague, Vaccine 20 (17–18)
[6] G. Kreil, Biosynthesis of melittin, a toxic peptide from bee venom. Amino-acid se- (2002) 2206–2214.
quence of the precursor, Eur. J. Biochem. 33 (3) (1973) 558–566. [38] T.O. Omotade, et al., D-cycloserine or similar physiochemical compounds may be

11
C.K. Cote, et al. Microbial Pathogenesis 142 (2020) 104050

uniquely suited for use in Bacillus anthracis spore decontamination strategies, J. [67] C. Kim, et al., Human alpha-defensins neutralize anthrax lethal toxin and protect
Appl. Microbiol. 115 (6) (2013) 1343–1356. against its fatal consequences, Proc. Natl. Acad. Sci. U. S. A. 102 (13) (2005)
[39] S.L. Welkos, et al., A microtiter fluorometric assay to detect the germination of 4830–4835.
Bacillus anthracis spores and the germination inhibitory effects of antibodies, J. [68] S. Welkos, et al., Humanized theta-defensins (retrocyclins) enhance macrophage
Microbiol. Methods 56 (2) (2004) 253–265. performance and protect mice from experimental anthrax infections, Antimicrob.
[40] CLSI, M7-A5 – Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria Agents Chemother. 55 (9) (2011) 4238–4250.
that Grow Aerobically, Clinical Laboratory Standards Institute, Wayne, PA, 2017. [69] J. Svenson, et al., Albumin binding of short cationic antimicrobial micropeptides
[41] H.S. Heine, et al., In vitro antibiotic susceptibilities of Yersinia pestis determined by and its influence on the in vitro bactericidal effect, J. Med. Chem. 50 (14) (2007)
broth microdilution following CLSI methods, Antimicrob. Agents Chemother. 59 (4) 3334–3339.
(2015) 1919–1921. [70] E.M. Molhoek, et al., Chicken cathelicidin-2-derived peptides with enhanced im-
[42] H.S. Heine, et al., In vitro antibiotic susceptibilities of Francisella tularensis de- munomodulatory and antibacterial activities against biological warfare agents, Int.
termined by broth microdilution following CLSI methods, Antimicrob. Agents J. Antimicrob. Agents 36 (3) (2010) 271–274.
Chemother. 61 (9) (2017). [71] D. Xhindoli, et al., The human cathelicidin LL-37–A pore-forming antibacterial
[43] M.E. Levison, Pharmacodynamics of antimicrobial drugs, Infect. Dis. Clin. North peptide and host-cell modulator, Biochim. Biophys. Acta 1858 (3) (2016) 546–566.
Am. 18 (3) (2004) 451–465 vii. [72] R.I. Lehrer, W. Lu, alpha-Defensins in human innate immunity, Immunol. Rev. 245
[44] M7-M5, Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that (1) (2012) 84–112.
Grow Aerobically, Clinical Laboratory Standards Institute, 2017. [73] L.R. Peterson, A review of tigecycline–the first glycylcycline, Int. J. Antimicrob.
[45] M.M. Sopirala, et al., Synergy testing by Etest, microdilution checkerboard, and Agents 32 (Suppl 4) (2008) S215–S222.
time-kill methods for pan-drug-resistant Acinetobacter baumannii, Antimicrob. [74] M. Linkevicius, L. Sandegren, D.I. Andersson, Potential of tetracycline resistance
Agents Chemother. 54 (11) (2010) 4678–4683. proteins to evolve tigecycline resistance, Antimicrob. Agents Chemother. 60 (2)
[46] P.J. Petersen, et al., In vitro antibacterial activities of tigecycline in combination (2016) 789–796.
with other antimicrobial agents determined by chequerboard and time-kill kinetic [75] D. Yahav, et al., Efficacy and safety of tigecycline: a systematic review and meta-
analysis, J. Antimicrob. Chemother. 57 (3) (2006) 573–576. analysis, J. Antimicrob. Chemother. 66 (9) (2011) 1963–1971.
[47] M.J. Hall, R.F. Middleton, D. Westmacott, The fractional inhibitory concentration [76] K. Smith, J.J. Leyden, Safety of doxycycline and minocycline: a systematic review,
(FIC) index as a measure of synergy, J. Antimicrob. Chemother. 11 (5) (1983) Clin. Therapeut. 27 (9) (2005) 1329–1342.
427–433. [77] R.K. Flamm, et al., In vitro activity of minocycline against U.S. Isolates of acine-
[48] D. Krogstad, R.C. Moellering, Antimicrobial combinations, Antibiotics in Laboratory tobacter baumannii-acinetobacter calcoaceticus species complex,
Medicine V. Lorian, Williams & Wilkins, Baltimore, MD, 1986, pp. 557–578. Stenotrophomonas maltophilia, and Burkholderia cepacia complex: results from the
[49] J.F. Acar, Antibiotic synergy and antagonism, Med. Clin. North Am. 84 (6) (2000) SENTRY antimicrobial surveillance program (2014-2018), Antimicrob. Agents
1391–1406. Chemother. 63 (2019), https://doi.org/10.1128/aac.01154-19 e01154-19.
[50] P.G. Spitzer, et al., Synergistic activity between vancomycin or teicoplanin and [78] N. Garrido-Mesa, A. Zarzuelo, J. Galvez, Minocycline: far beyond an antibiotic, Br.
gentamicin or tobramycin against pathogenic diphtheroids, Antimicrob. Agents J. Pharmacol. 169 (2) (2013) 337–352.
Chemother. 32 (4) (1988) 434–437. [79] G.S. Bisacchi, J.I. Manchester, A new-class Antibacterial-almost. Lessons in drug
[51] J. Li, et al., Antimicrobial activity and resistance: influencing factors, Front. discovery and development: a critical analysis of more than 50 Years of effort to-
Pharmacol. 8 (2017) 364. ward ATPase inhibitors of DNA gyrase and topoisomerase IV, ACS Infect. Dis. 1 (1)
[52] M.L. Pato, Tetracycline inhibits propagation of deoxyribonucleic acid replication (2015) 4–41.
and alters membrane properties, Antimicrob. Agents Chemother. 11 (2) (1977) [80] T. Khan, et al., DNA gyrase inhibitors: progress and synthesis of potent compounds
318–323. as antibacterial agents, Biomed. Pharmacother. 103 (2018) 923–938.
[53] M.A. Kohanski, D.J. Dwyer, J.J. Collins, How antibiotics kill bacteria: from targets [81] F.M. Thibault, et al., Antibiotic susceptibility of 65 isolates of Burkholderia pseu-
to networks, Nat. Rev. Microbiol. 8 (6) (2010) 423–435. domallei and Burkholderia mallei to 35 antimicrobial agents, J. Antimicrob.
[54] M.S. Zharkova, et al., Application of antimicrobial peptides of the innate immune Chemother. 54 (6) (2004) 1134–1138.
system in combination with conventional antibiotics-A novel way to combat anti- [82] F. Tonello, I. Zornetta, Bacillus anthracis factors for phagosomal escape, Toxins
biotic resistance? Front. Cell Infect. Microbiol. 9 (2019) 128. (Basel) 4 (7) (2012) 536–553.
[55] S. Leekha, C.L. Terrell, R.S. Edson, General principles of antimicrobial therapy, [83] Y. Ke, Z. Chen, R. Yang, Yersinia pestis: mechanisms of entry into and resistance to
Mayo Clin. Proc. 86 (2) (2011) 156–167. the host cell, Front. Cell Infect. Microbiol. 3 (2013) 106.
[56] P.D. Tamma, S.E. Cosgrove, L.L. Maragakis, Combination therapy for treatment of [84] J. Celli, T.C. Zahrt, Mechanisms of Francisella tularensis intracellular pathogenesis,
infections with gram-negative bacteria, Clin. Microbiol. Rev. 25 (3) (2012) Cold Spring Harb. Perspect. Med. 3 (4) (2013) a010314.
450–470. [85] L. Whiteley, et al., Entry, intracellular survival, and multinucleated-giant-cell-
[57] R. Guner, et al., Outcomes in patients infected with carbapenem-resistant forming activity of Burkholderia pseudomallei in human primary phagocytic and
Acinetobacter baumannii and treated with tigecycline alone or in combination nonphagocytic cells, Infect. Immun. 85 (10) (2017).
therapy, Infection 39 (6) (2011) 515–518. [86] G.C. Whitlock, et al., Burkholderia mallei cellular interactions in a respiratory cell
[58] J. Lee, et al., Decreased susceptibility to polymyxin B during treatment for carba- model, J. Med. Microbiol. 58 (Pt 5) (2009) 554–562.
penem-resistant Klebsiella pneumoniae infection, J. Clin. Microbiol. 47 (5) (2009) [87] K. Matsuzaki, Control of cell selectivity of antimicrobial peptides, Biochim. Biophys.
1611–1612. Acta 1788 (8) (2009) 1687–1692.
[59] B. Deslouches, Y.P. Di, Antimicrobial peptides: a potential therapeutic option for [88] D. Takahashi, et al., Structural determinants of host defense peptides for anti-
surgical site infections, Clin. Surg. 2 (2017). microbial activity and target cell selectivity, Biochimie 92 (9) (2010) 1236–1241.
[60] S.T. Yang, S.Y. Shin, J.I. Kim, Interaction mode of a symmetric Trp-rich undeca [89] Safety Data Sheet-Dimethyl sulfoxide (DMSO). Sigma Aldrich, Version 6.4, Revised
peptide PST11-RK with lipid bilayers, FEBS Lett. 581 (1) (2007) 157–163. 01-15-20.
[61] M. Mardirossian, et al., The host antimicrobial peptide Bac71-35 binds to bacterial [90] Q. Guo, et al., Potential use of dimethly sulfoxide in treatment of infections caused
ribosomal proteins and inhibits protein synthesis, Chem. Biol. 21 (12) (2014) by Pseudomonas aeruginosa, Antimicrob. Agents Chemother. 60 (12) (2016)
1639–1647. 7159–7169.
[62] L.T. Nguyen, E.F. Haney, H.J. Vogel, The expanding scope of antimicrobial peptide [91] S.W. Shirley, B.H. Stewart, S. Mirelman, Dimethyl sulfoxide in treatment of in-
structures and their modes of action, Trends Biotechnol. 29 (9) (2011) 464–472. flammatory genitourinary disorders, Urology 11 (3) (1978) 215–220.
[63] L.P. Lashua, et al., Engineered cationic antimicrobial peptide (eCAP) prevents [92] E.A. Thackaberry, et al., Solvent-based formulations for intravenous mouse phar-
Pseudomonas aeruginosa biofilm growth on airway epithelial cells, J. Antimicrob. macokinetic studies: tolerability and recommended solvent dose limits, Xenobiotica
Chemother. 71 (8) (2016) 2200–2207. 44 (3) (2014) 235–241.
[64] Y. Huang, et al., Cationic amphiphilic alpha-helical peptides for the treatment of [93] E.G. Worthley, C.D. Schott, The toxicity of four concentrations of DMSO, Toxicol.
carbapenem-resistant Acinetobacter baumannii infection, Biomaterials 33 (34) Appl. Pharmacol. 15 (1969) 275–281.
(2012) 8841–8847. [94] B. Deslouches, et al., De novo-derived cationic antimicrobial peptide activity in a
[65] M.L. Mangoni, et al., Comparative analysis of the bactericidal activities of amphi- murine model of Pseudomonas aeruginosa bacteraemia, J. Antimicrob. Chemother.
bian peptide analogues against multidrug-resistant nosocomial bacterial strains, 60 (2007) 669–672.
Antimicrob. Agents Chemother. 52 (1) (2008) 85–91. [95] B. Deslouches, et al., Engineered cationic antimicrobial peptides to overcome
[66] A. Peschel, H.G. Sahl, The co-evolution of host cationic antimicrobial peptides and multidrug resistance by ESKAPE pathogens, Antimicrob. Agents Chemother. 59 (2)
microbial resistance, Nat. Rev. Microbiol. 4 (7) (2006) 529–536. (2015) 1329–1333.

12

You might also like