AAC Accepted Manuscript Posted Online 8 February 2016 Antimicrob. Agents Chemother. doi:10.1128/AAC.02972-15 Copyright © 2016, American Society for Microbiology. All Rights Reserved.

1

Novel engineered peptides of a phage lysin as effective antimicrobials against multidrug

2

resistant Acinetobacter baumannii

3

Mya Thandar1§, Rolf Lood1,2§, Benjamin Y. Winer1,3§, Douglas R. Deutsch1, Chad W.

4

Euler1,4,5, Vincent A. Fischetti1#

5

1

6

York Avenue, New York, NY 10065, USA

7

2

8

Lund University, Sweden

9

3

Laboratory of Bacterial Pathogenesis and Immunology, The Rockefeller University, 1230

Present address: Department of Clinical Sciences Lund, Division of Infection Medicine,

Present address: 119 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey,

10

NJ, 08544-1014, USA

11

4

12

Hunter College, CUNY, New York, NY, 10065, United States of America.

13

5

14

College, New York, NY, 10065, United States of America.

15

Short title: Antibacterial peptides active against Acinetobacter baumannii

16

# Corresponding author: Vincent A. Fischetti, [email protected]

17

§

Present address: Department of Medical Laboratory Sciences, Belfer Research Building,

Present address: Department of Microbiology and Immunology, Weill Cornell Medical

These authors contributed equally.

18 19

1

20

Abstract

21

Acinetobacter baumannii is a Gram-negative bacterial pathogen responsible for a range of

22

nosocomial infections. The recent rise and spread of multidrug resistant A. baumannii clones

23

has fueled a search for alternative therapies, including bacteriophage endolysins with potent

24

antibacterial activities. A common feature of these lysins is the presence of a highly

25

positively charged C-terminal domain with a likely role in promoting outer membrane

26

penetration. In the current study, we show that the C-terminal amino acid 108-138 of phage

27

lysin PlyF307, named P307, alone was sufficient to kill A. baumannii (>3-logs). Furthermore,

28

P307 could be engineered for improved activity, the most active derivative being P307SQ-8C

29

(>5-log kill). Both P307 and P307SQ-8C showed high in vitro activity against A. baumannii in

30

biofilms. Moreover, P307SQ-8C exhibited MICs comparable to levofloxacin and ceftazidime

31

and acted synergistically with polymyxin B. While the peptides were shown to kill by

32

disrupting the bacterial cytoplasmic membrane, they did not lyse human red blood cells or B

33

cells; however, serum was found to be inhibitory to lytic activity. In a murine model of A.

34

baumannii skin infection, P307SQ-8C reduced the bacterial burden by ~2-logs in 2 h. This

35

study demonstrates the prospect of using peptide derivatives from bacteriophage lysins to

36

treat topical infections and remove biofilms caused by Gram-negative pathogens.

37

Introduction

38

Acinetobacter baumannii is an increasingly significant nosocomial pathogen worldwide (1).

39

Arising from both intrinsic and acquired antibiotic resistance, multi- and pan-drug resistant

40

clones of A. baumannii can readily be isolated from hospital environments (2). A. baumannii

41

has been shown to develop resistance to several classes of antibiotics, including

42

aminoglycosides, cephalosporins, carbapenems, tigecycline and colistin (3). The reasons for

43

high resistance include a high degree of genetic plasticity combined with an intrinsic

2

44

resistance to certain antibiotics due to the presence of β-lactamases, low permeability of the

45

outer membrane, and highly efficient efflux pump systems (4). Furthermore, A. baumannii is

46

prone to develop biofilms on solid surfaces, including medical devices (5). Thus, A.

47

baumannii is not only problematic as an infectious agent, but it is also increasingly difficult

48

to remove from hospital environments – a phenomenon similar to the Gram-positive

49

nosocomial pathogen Staphylococcus aureus.

50

One of the last resort antibiotics for A. baumannii is the antimicrobial peptide polymyxin B

51

(6). The bactericidal effect of polymyxin B is mediated through its positively charged Dab

52

(α,γ-diaminobutyric acid) residues interacting with LPS and destabilizing the outer

53

membrane (7). Many antimicrobial peptides kill in a similar way: clustered cationic residues

54

permeabilize the bacterial membrane to cause lysis and death (8). Due to this mechanism of

55

action, most of the membrane-acting antimicrobial peptides commonly have cytotoxic effects

56

on eukaryotic cells (9). Indeed, polymyxin B has severe side effects: cytotoxicity,

57

nephrotoxicity and neurotoxicity (10). Since careful administration is required to avoid its

58

toxicity, the dose range of polymyxin B is limited and resistant strains of A. baumannii have

59

been documented (11).

60

Recently, there has been a growing interest in the use of bacterial viruses (i.e., bacteriophage

61

therapy) to treat infections by Gram-negative bacteria, including A. baumannii (12-14).

62

Several phages that infect A. baumannii have been identified and characterized. However,

63

their restricted spectrum (killing only ∼60% of A. baumannii isolates) limits the effectiveness

64

of such phages as therapeutic agents (12, 13). Using an alternative bacteriophage-based

65

approach, our group and others have taken advantage of the lytic enzymes (lysins) encoded

66

and produced by bacteriophages during lytic proliferation (15-18). Bacteriophage lysins are

67

classified as peptidoglycan hydrolases, being able to cleave a variety of bonds in the bacterial

3

68

peptidoglycan. Cleavage of the cell wall by lysins destabilizes the peptidoglycan and

69

weakens the structural framework, resulting in hypotonic lysis. While purified lysins are

70

effective at killing Gram-positive bacteria (19), the outer membrane of Gram-negative

71

bacteria largely limits lysins from accessing and cleaving the subjacent peptidoglycan.

72

Different strategies have been employed to increase the efficiency of lysins against Gram-

73

negative bacteria, including the use of the chelating agent EDTA (16, 17), and the genetic

74

engineering of lysins to add either highly charged/hydrophobic N-/C-terminal extensions (20)

75

or other membrane translocating domains (21, 22). However, there has been little focus on

76

the intrinsic features of certain active lysins against Gram-negative bacteria, and how they

77

function to allow the lysins to cross the outer membrane and reach the subjacent

78

peptidoglycan substrate.

79

Here, we have identified a highly cationic C-terminal domain within an A. baumannii phage

80

lysin as a peptide with potent antibacterial activity. We have modified the peptide to further

81

improve its activity, and have proven the high efficiency of such peptides to kill A.

82

baumannii both in vitro and in vivo in a skin infection model.

83

Materials and Methods

84

Bacterial strains and growth conditions

85

Acinetobacter baumannii strains in this study include clinical isolates from Hospital for

86

Special Surgery in New York (#1775-#1799) (18), Ohio State University (S1, S3 and S5

87

provided by Dr. Vijay Pancholi), and ATCC® 17978™ from American Type Culture

88

Collection. Bacteria were cultured in Trypticase Soy Broth or Brain Heart Infusion (TSB or

89

BHI; Thermo Fisher Scientific, Waltham, MA, USA) at 37°C with aeration (200 rpm).

90

Stationary phase bacteria were cultured overnight while log phase bacteria were grown for 3

4

91

h in fresh media from 100x dilutions of overnight cultures. Strains for determining the

92

specificity of the antimicrobial peptides were cultured under the same conditions, at the

93

temperatures indicated: Bacillus anthracis ΔSterne (30°C), Escherichia coli DH5α (37°C),

94

Klebsiella pneumoniae ATCC® 700603TM and 10031TM (37°C), Pseudomonas aeruginosa

95

PAO1 (30°C), and Staphylococcus aureus RN4220 (30°C).

96

Biofilms were set up as previously described (18). Briefly, an overnight culture of A.

97

baumannii strain #1791 was diluted 1000x (~105 cfu/mL) in TSB containing 0.2% glucose

98

and incubated at 37°C for 72 h in ~2.5 cm segments of PVC catheter tubing (CareFusion)

99

with the ends clamped shut to prevent evaporation. The catheter biofilms were washed with

100

autoclaved Milli-Q water to remove planktonic cells before peptide treatment. Crystal violet

101

(0.1%) was used to detect the presence of biofilm.

102

Peptide synthesis and preparation

103

Peptides were synthesized at The Rockefeller University Proteomics Resource Center. All

104

peptides were created using a Protein Technologies Symphony™ peptide synthesizer (PTI

105

Tucson, Arizona, USA) on pre-coupled Wang (p-alkoxy-benzyl alcohol) resin (Bachem,

106

Torrance, CA, USA). Reaction vessels were loaded at 25 μM and peptides were elongated

107

using Fmoc protected amino acids (Anaspec, San Jose, CA, USA) (23). Deprotection of the

108

amine was accomplished with 20% piperidine (Sigma-Aldrich) in NMP (N-

109

methylpyrrolidinone). Repetitive coupling reactions were conducted using 0.6 M HATU/Cl-

110

HOBT (azabenzotriazol tetramethyluronium hexafluorophosphate/6-chloro-1-

111

hydroxybenzotriazole)(P3 Biosystems, Shelbyville, KY, USA) and 0.4 M NMM (N-

112

methylmorpholine) using NMP (EMD) as the primary solvent (24). Resin cleavage and side-

113

chain deprotection were achieved by transferring to a 100 mL round bottom flask and reacted

114

with 4.0 mL concentrated, sequencing grade, trifluoracetic acid (Fisher) with 5

115

triisopropylsilane (Fluka), degassed water, and 3,6-dioxa-1,8-octanedithiol (DODT, Sigma-

116

Aldrich) in a ratio of 95:2:2:1 over a 6 h time frame. This was followed by column filtration

117

to a 50 mL round bottom flask and TFA volume reduced to 2 mL using a rotary evaporator.

118

A standard ether precipitation was performed on the individual peptides by transferring to a

119

50 mL falcon tube containing 40 mL cold tert-butyl methyl ether (TBME, Sigma-Aldrich).

120

Samples were placed in an ice bath for 2 h to aid precipitation followed by pellet formation

121

using centrifugation (3300 rpm, 5 min). Excess ether was removed by vacuum aspiration and

122

the peptide pellets were allowed to dry overnight in a fume hood. Dried peptide pellets were

123

resolved in 20% acetonitrile and 10 mL HPLC grade water, subsampled for LC/MS and

124

lyophilized. All crude products were subsequently analyzed by reverse-phase Aquity™

125

UPLC (Waters Chromatography, Milford, MA, USA) using a Waters BEH C18 column.

126

Individual peptide integrity was verified by tandem electrospray mass spectrometry using a

127

ThermoFinnigan LTQ™ (Thermo Fisher, Waltham, MA, USA) spectrometer system.

128

Preparative chromatography was accomplished on a Vydac C18 RP preparative column on a

129

Waters 600 Prep HPLC. Individual fractions were collected in 30 s intervals, characterized

130

using LC/MS and fractions containing desired product were lyophilized. These were stored at

131

-20°C until being re-suspended in autoclaved Milli-Q water for various assays. The stock

132

solutions were then stored at 4°C.

133

Bactericidal assays

134

Unless otherwise indicated, stationary phase bacteria (overnight cultures) were used for the

135

assays. Bacteria were washed in assay buffer, re-suspended to ∼106 cfu/mL, mixed with each

136

antimicrobial peptide and incubated for 2 h at room temperature (22-25°C). After treatment,

137

the reactions were serially diluted and plated for enumeration. Influencing factors on the

138

activities of the peptides (P307 and P307SQ-8C) were examined: buffer (50 mM sodium

6

139

phosphate or Tris-HCl, pH 6.8-8.8; NaCl (0-200 mM); concentration (0-125 μg/mL P307);

140

time (1-120 min); specificity (the bacterial strains mentioned above); and growth phase (log,

141

stationary and biofilm). The buffer system for the experiments with the latter five factors was

142

50 mM Tris-HCl, pH 7.5. The biofilm-associated bacteria were re-suspended by thoroughly

143

scraping the catheter tubing and vortexing for 1 min.

144

To examine the mechanism(s) of action of the peptides, the following bactericidal assays

145

were also conducted. The contribution of disulfide bond formation to the increased activity of

146

P307SQ-8C was investigated by comparing with P307SQ-8A in the presence of the reducing

147

agent TCEP (0.1 and 1 mM) (Tris (2-carboxyethyl) phosphine hydrochloride

148

solution)(Sigma). To examine the importance of ionic interaction, the least sensitive Gram-

149

negative bacterial species at pH 7.5 were treated with P307 and P307SQ-8C at pH 8.8.

150

Experiments were conducted at least in triplicate and representative data are shown as mean

151

± standard deviation. The black horizontal lines mark the limit of detection.

152

Minimum inhibitory concentrations (MICs) and synergy

153

The broth microdilution method was used (25) to determine the MICs of levofloxacin,

154

ceftazidime, polymyxin B, P307 and P307SQ-8C for A. baumannii strains #1791, #1798, S5

155

and ATCC17978. For the antibiotics, 1.5–2 fold serial dilutions (three lower and three

156

higher) of the MICs determined by Etest (18) were included. For the peptides, two-fold serial

157

dilutions (2000–31.25 μg/mL) were tested. Overnight cultures were re-suspended to ~105

158

cfu/mL in Mueller Hinton broth (pH 7.9). Antibiotics or peptides were added to a final

159

volume of 100 μL for each dilution. Bacteria were allowed to grow at 37°C for 24 h at 220

160

rpm. The absorbance at 595 nm was then read in a SpectraMax Plus Reader (Molecular

161

Devices). The MICs were determined as the lowest concentrations of antimicrobial agents

7

162

that visibly inhibited bacterial growth. In addition, Alamar®Blue was used to confirm the

163

data obtained from OD595. Each experiment was repeated at least twice in duplicate.

164

To investigate whether synergy exists between each peptide and antibiotic, checkerboard

165

assays were conducted by mixing fractional inhibitory concentrations (FICs) of antibiotics

166

(levofloxacin, ceftazidime and polymyxin B) with those of peptides P307 and P307SQ-8C as

167

previously described (26). Isobolograms were constructed to determine the fractional

168

inhibitory concentration indices (FICIs). Each experiment was repeated at least three times.

169

DNA binding assay

170

The affinity of peptide P307 for DNA was investigated by electrophoretic mobility shift

171

assay (27). P307 was mixed with 50 ng of random DNA (1 kb amplicon of Bdellovibrio

172

genomic DNA) at different peptide to DNA ratios (0:1-15:1) in a binding buffer (5 mM Tris-

173

HCl, pH 8.0, 10 mM EDTA, 5% glycerol, 20 mM KCl, 50 μg/mL BSA). As a positive

174

control, a peptide from Bdellovibrio bacteriovorus that was previously found in our

175

laboratory to have DNA-binding capacity (amino acid sequence –

176

MASKTKKTEYIRERKKATSGKKRKAANRTKGTTKSAKTLFKD) was used

177

(unpublished results). After incubation for 1 h at 22-25°C, the peptide and DNA mixture was

178

analyzed by agarose gel electrophoresis (27).

179

Transmission electron microscopy (TEM)

180

An overnight culture of A. baumannii strain #1791 was collected, washed in 50 mM Tris-HCl,

181

pH 7.5 and re-suspended in the same buffer to ~108 cfu/mL. The bacteria were treated with

182

300 μg/mL P307SQ-8C or buffer (control) for 5 min or 2 h. The samples were fixed with 2.5%

183

glyceraldehyde in 0.1 M sodium cacodylate, and then TEM images were taken at

184

magnification ×2600 or ×5000. Representative figures are shown. 8

185

SYTOX Green uptake assay

186

The permeability of bacterial membrane upon peptide treatment was measured by SYTOX®

187

Green uptake (28). Briefly, overnight cultures of bacteria were washed and re-suspended to

188

∼107 cfu/mL in 50 mM Tris-HCl pH 7.5. Benzonase® nuclease (25 U/mL)(Novagen) and

189

SYTOX® Green (1 μM)(Invitrogen) was added to the bacterial cells, and incubated for 15

190

min at 22-25°C in the dark. Peptides (50 μg/mL) were added, and polymyxin B (2

191

μg/mL)(Sigma) was used as a control (29). Relative fluorescence units (RFU) were measured

192

in a SpectraMax Plus reader (Molecular Devices) at 22-25°C (ex: 485 nm, em: 520 nm) for 2

193

h. Experiments were carried out twice in duplicate and representative data are shown as mean

194

± standard deviation.

195

Cytotoxicity assays

196

The hemolytic assays were conducted as previously described (28). Briefly, human blood

197

was gathered in an EDTA-tube, and red blood cells (RBC) were collected through low speed

198

centrifugation. Cells were washed and re-suspended to a 10% RBC solution in PBS, and

199

mixed with 345 μg/mL P307SQ-8C. PBS and 1% Triton X-100 were used as negative and

200

positive controls, respectively. After 1 h incubation at 37°C, the supernatant was collected,

201

and the absorbance at 405 nm was recorded through SpectraMax Plus Reader (Molecular

202

Devices) to measure the release of hemoglobin. The reactions were carried out twice in

203

triplicate and representative data are shown as mean ± standard deviation.

204

A human B lymphoblastoid cell line (LCL) obtained from The Rockefeller University was

205

grown in RPMI media supplemented with L-glutamine and 10% fetal bovine serum. Cells

206

were harvested by low speed centrifugation, washed and re-suspended in PBS to ~5x106 live

207

cells/mL, as determined by trypan blue exclusion tests. LCL cells (~5x105) were incubated

9

208

with 172.5, 345 and 517.5 μg/mL P307SQ-8C at 37°C in a humidified 5% CO2 atmosphere for

209

one hour. The cells were then stained according to manufacturer’s instructions (CellTiter 96

210

Non-radioactive cell proliferation assay, Promega) where live B cells reduced the dye to

211

insoluble formazan for an additional 4 h. Solubilization/Stop solution was added and

212

incubated at 37°C overnight. Next, the absorbance at 570 nm was measured in SpectraMax

213

Plus Reader (Molecular Devices) to quantitate B cell survival. Triton X-100 was used as a

214

positive control. The reactions were carried out in triplicate and data are shown as mean ±

215

standard deviation.

216

Endotoxin release

217

To detect the amount of endotoxin release, strain #1791 (~107 cfu/mL) was incubated at 22-

218

25°C for 2 h with different concentrations of P307 or P307SQ-8C. The samples were briefly

219

centrifuged (2000 × g) for 1 min to remove live cells. All reagents were prepared in

220

endotoxin-free autoclaved Milli-Q water. Endpoint Chromogenic LAL Assay (Lonza) was

221

conducted according to manufacturer’s instructions. Polymyxin B and 100% ethanol were

222

used for comparison and positive control, respectively. The experiment was conducted twice

223

in duplicate and representative data are shown as mean ± standard deviation.

224

Bactericidal activity in plasma and its components

225

Human blood was collected in a heparin-tube and centrifuged. The supernatant was filtered

226

and stored at 4°C overnight. The filtrate was centrifuged and filtered again to remove any

227

debris. The resulting solution was used as 100% plasma. The components of plasma

228

examined for interference were monovalent cation (Na+), divalent cations (Ca2+ and Mg2+)

229

and albumin. Chloride salts of the cations and albumin from human serum (lyophilized

230

powder, ≥ 97%)(Sigma) were prepared in Milli-Q water and sterile filtered. Bactericidal

10

231

assays, as described above, were conducted in 100% plasma, and in buffered solutions (50

232

mM Tris-HCl, pH 7.5) containing 150 mM NaCl, 1 mM CaCl2, 1 mM MgCl2, and 50 mg/mL

233

human serum albumin (HSA), using 100 µg/mL P307 or P307SQ-8C. The experiments were

234

conducted at least in triplicate and data are shown as mean ± standard deviation.

235

Mouse in vivo skin model

236

The Rockefeller University institutional animal care and use committee approved all in vivo

237

protocols. Skin infection was induced with A. baumannii as previously described (30).

238

Briefly, the backs of 30 female CD-1 mice (6 to 8 weeks of age; Charles River Laboratories)

239

were shaved with an electric razor. NairTM depilatory cream was applied to the shaved areas

240

to remove any remaining hair. The areas were then disinfected with alcohol wipes, and tape

241

stripped with autoclave tape 20 times in succession, using a fresh piece of tape each time.

242

The tape stripped areas were disinfected again with alcohol wipes. An area of ~ 1 cm2 was

243

then marked and infected with 10 μL of ~108 cfu/mL A. baumannii strain #1791. The bacteria

244

were allowed to colonize for 16-18 h, after which the infected area was either left untreated

245

or treated with 200 μg of P307SQ-8C or 2 μg of polymyxin B in autoclaved Milli-Q water for 2

246

h. To harvest the remaining bacteria on the skin, the mice were sacrificed and the infected

247

skin was processed in 500 μL PBS for 1-2 min in a Stomacher® 80 Biomaster using a

248

microbag (Seward Ltd., UK). The solution was serially diluted and plated on LB agar

249

containing 4 μg/mL levofloxacin and 12 μg/mL ampicillin for selection. The resulting

250

cfu/mL from each animal was plotted as an individual point and the horizontal bars represent

251

the mean values. Data were analyzed using Ordinary one-way ANOVA in GraphPad Prism

252

6.0. The dotted horizontal line marks the limit of detection.

253

Results

11

254

Identification and modification of putative antibacterial peptides from phage lysin PlyF307

255

Previously published works on phage lysins against Gram-negative bacteria (15, 18)

256

suggested that the C-terminal portions of lysins might destabilize the bacterial outer

257

membrane to facilitate access to the lysin’s substrate, i.e., the peptidoglycan. Specifically, the

258

highly cationic C-terminal region of A. baumannii phage lysin LysAB2 was speculated to be

259

involved in permeabilizing the outer membrane (15). We were able to identify a similar

260

highly positively charged sequence in one of our A. baumannii phage lysins, PlyF307 (18).

261

This peptide, termed P307 (Table 1), extends from amino acids 108-138 in PlyF307, and

262

partly resembles the LysAB2 peptide in terms of length and charge, but sharing only 41.2%

263

similarity and 17.6% identity (MacVector ClustalW alignment).

264

We modified P307 with the eight amino acids constituting the full C-terminal part of native

265

PlyF307, forming peptide P307AE-8 (Table 1). In addition, we fused eight amino acids

266

(SQSRESQC) to the C-terminus of P307 and yielded P307SQ-8C (Table 1). This modification

267

was based on the observation of an earlier study showing that the lack of these eight amino

268

acids reduces the activity spectrum of the antimicrobial peptide from Hepatitis B virus core

269

protein (28). We also generated two additional modifications of P307SQ-8C: scrambling the

270

sequence of the last eight amino acids to CSQRQSES (P307CS-8); and changing the last

271

amino acid from C to A (P307SQ-8A) (Table 1). The sequence was scrambled to evaluate the

272

importance of a specific sequence and the cysteine change was introduced to examine the

273

importance of intermolecular disulfide bond formation for activity. A predicted structure of

274

PlyF307, generated through I-TASSER (31-33), suggested that P307 and its derivatives

275

formed a “hairpin-like” di-alpha helical structure connected with a flexible linker region (Fig.

276

1).

277

Comparison of the peptides 12

278

We evaluated the bactericidal activities of the peptides P307, P307AE-8 and P307SQ-8C.

279

P307SQ-8C was the most active, reducing ~106 cfu/mL of bacteria to below the limit of

280

detection (5-

323

log decrease). Biofilm-associated A. baumannii was more resistant (Fig. 4B). Biofilms were

324

treated with 250 μg/mL P307 or P307SQ-8C for 2 or 24 h. After 2 h, ~3- and 4-log decrease in

14

325

cfu/mL was observed with P307 and P307SQ-8C, respectively. After 24 h, an additional ~1.3-

326

log decrease was observed with P307 while there was no further decrease with P307SQ-8C.

327

Minimal inhibitory concentration and synergy

328

In order to compare the efficiency of the peptides with clinically used antibiotics, we

329

performed minimal inhibitory concentration (MIC) assays on four A. baumannii strains:

330

#1791, #1798, S5 and ATCC17978. The strains displayed varying degrees of sensitivity to

331

levofloxacin, ceftazidime and P307 while the MICs of polymyxin B and P307SQ-8C were more

332

consistent (Table 2). P307SQ-8C had a lower MIC than P307, which was in accordance with in

333

vitro killing activity (Fig. 2 and 3).

334

We also examined the synergistic effects between each peptide and levofloxacin, ceftazidime

335

or polymyxin B by the checkerboard method (isobologram). No synergy was observed

336

between either peptide and levofloxacin or ceftazidime (data not shown). However,

337

polymyxin B acted synergistically with both P307 and P307SQ-8C (FICIs = 0.125, 1024 >1873

0.25 0.19

1000 294

62.5 14.5

6 16.6

64 117

0.25 0.19

2000 588

125 29

6 11

0.25 0.19

750 221

125 29

S5 ATCC17978 616

4.3

0.075 0.21

Ceftazidime

Polymyxin B

P307

P307SQ-8C

µg/mL µM µg/mL µM

a

Numbers in bold indicate resistance to the antibiotics.

26

Novel Engineered Peptides of a Phage Lysin as Effective Antimicrobials against Multidrug-Resistant Acinetobacter baumannii.

Acinetobacter baumannii is a Gram-negative bacterial pathogen responsible for a range of nosocomial infections. The recent rise and spread of multidru...
950KB Sizes 0 Downloads 10 Views