29
Accepted Manuscript A novel tool against multiresistant bacterial pathogens – lipopeptide modification of the natural antimicrobial peptide ranalexin for enhanced antimicrobial activity and improved pharmacokinetics Cornelius Domhan , Philipp Uhl , Anna Meinhardt , Stefan Zimmermann , Christian Kleist , Thomas Lindner , Karin Leotta , Walter Mier , Michael Wink PII: S0924-8579(18)30101-8 DOI: 10.1016/j.ijantimicag.2018.03.023 Reference: ANTAGE 5414 To appear in: International Journal of Antimicrobial Agents Received date: 16 November 2017 Revised date: 29 March 2018 Accepted date: 31 March 2018 Please cite this article as: Cornelius Domhan , Philipp Uhl , Anna Meinhardt , Stefan Zimmermann , Christian Kleist , Thomas Lindner , Karin Leotta , Walter Mier , Michael Wink , A novel tool against multiresistant bacterial pathogens – lipopeptide modification of the natural antimicrobial peptide ranalexin for enhanced antimicrobial activity and improved pharmacokinetics, International Journal of Antimicrobial Agents (2018), doi: 10.1016/j.ijantimicag.2018.03.023 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

Accepted Manuscript

A novel tool against multiresistant bacterial pathogens – lipopeptidemodification of the natural antimicrobial peptide ranalexin forenhanced antimicrobial activity and improved pharmacokinetics

Cornelius Domhan , Philipp Uhl , Anna Meinhardt ,Stefan Zimmermann , Christian Kleist , Thomas Lindner ,Karin Leotta , Walter Mier , Michael Wink

PII: S0924-8579(18)30101-8DOI: 10.1016/j.ijantimicag.2018.03.023Reference: ANTAGE 5414

To appear in: International Journal of Antimicrobial Agents

Received date: 16 November 2017Revised date: 29 March 2018Accepted date: 31 March 2018

Please cite this article as: Cornelius Domhan , Philipp Uhl , Anna Meinhardt , Stefan Zimmermann ,Christian Kleist , Thomas Lindner , Karin Leotta , Walter Mier , Michael Wink , A novel tool againstmultiresistant bacterial pathogens – lipopeptide modification of the natural antimicrobial peptideranalexin for enhanced antimicrobial activity and improved pharmacokinetics, International Journalof Antimicrobial Agents (2018), doi: 10.1016/j.ijantimicag.2018.03.023

This is a PDF file of an unedited manuscript that has been accepted for publication. As a serviceto our customers we are providing this early version of the manuscript. The manuscript will undergocopyediting, typesetting, and review of the resulting proof before it is published in its final form. Pleasenote that during the production process errors may be discovered which could affect the content, andall legal disclaimers that apply to the journal pertain.

Page 2: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Highlights

The antimicrobial peptide ranalexin showed strong antimicrobial activity against Gram-

positive bacteria and Acinetobacter species.

Ranalexin is excreted via the kidneys within one hour in WISTAR rats.

Lipopeptide derivatives of ranalexin displayed enhanced antimicrobial activity, especially

against Gram-negative bacteria.

The investigated lipopeptide derivative accumulated in the liver of WISTAR rats.

Time-kill studies showed a fast concentration-dependent killing within one hour.

Cytotoxicity of the lipopeptides depends on the chain length of the fatty acid.

Page 3: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

A novel tool against multiresistant bacterial pathogens – lipopeptide modification of the natural

antimicrobial peptide ranalexin for enhanced antimicrobial activity and improved

pharmacokinetics

Cornelius Domhan1, Philipp Uhl2, Anna Meinhardt², Stefan Zimmermann3, Christian Kleist2, Thomas

Lindner2, Karin Leotta2, Walter Mier2, Michael Wink1*

1 Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, 69120 Heidelberg,

Germany 2 Department of Nuclear Medicine, Heidelberg University Hospital, 69120 Heidelberg, Germany 3 Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University

Hospital, 69120 Heidelberg, Germany

* Corresponding author

Key words

Ranalexin, Antimicrobial peptides, Lipopeptides, Positron emission tomography, Multiresistant

bacteria, Drug development

Abstract

As evolutionary optimized defense compounds, antimicrobial peptides (AMPs) represent a powerful

tool against bacterial infections. Ranalexin, an AMP found in the skin of the American bullfrog (Rana

catesbeiana), is primarily active against Gram-positive bacteria with a minimum inhibitory

concentration (MIC) of 8 – 16 mg/L but shows weaker activity against Gram-negative bacteria (> 64

mg/L). By substitution of 6 N-terminal amino acids by decanoic (C10:0) to myristic (C14:0) acid, we

were able to develop lipopeptide derivatives with enhanced antimicrobial activity. The antimicrobial

capacity of the peptides was tested against different bacterial strains, including multiresistant clinical

isolates. C13C3lexin, the most potent derivative showed MICs of 2 – 8 mg/L against Gram-positive

bacteria and 2 – 16 mg/L against Gram-negative bacteria. In time-kill studies, it could be clearly

shown that ranalexin and the lipopeptide C13C3lexin function as concentration dependent, fast

acting substances against different bacteria. Cell viability assays revealed that cytotoxicity towards

human cells increases with the chain length of the attached fatty acid (IC50 13,65 – 108,10 µg/mL).

Furthermore, with positron emission tomography (PET)-imaging pharmacokinetics studies of 68Ga-

labeled ranalexin and its derivatives were performed for the first time. Here we could demonstrate

that ranalexin is rapidly cleared via the kidneys within 1 h post injection. In contrast, the lipopeptide

Page 4: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

showed strongly extended circulation within the bloodstream and a shift from renal to hepatic

accumulation characteristics. Therefore, the more favorable pharmacokinetics and enhanced

antimicrobial activity clearly demonstrate the potential of the lipopeptidic AMPs as novel bullets

against emerging multiresistant bacterial pathogens.

Page 5: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

1. Introduction

Prior to the introduction of antibiotics into medicine about 70 years ago, bacterial infections

represented a major cause of death. Unfortunately, antibiotic resistance developed and is now

spreading faster than the launch of new effective antimicrobial drugs into the market, potentially

leading to a health crisis in the near future [1]. Nowadays, increasing numbers of infections occur

caused by bacteria which are resistant to most of the antibacterial treatments currently available [2].

It is estimated that more than 2,000,000 illnesses and 23,000 deaths are caused by antibiotic

resistance in the United States per year [3]. The majority of severe infections are caused by bacteria,

generally referred to as “ESKAPE”, comprising Enterococcus faecium, Staphylococcus aureus,

Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter species

[4]. Infections caused by the multidrug resistant A. baumannii belong to infections most difficult to

treat [5].

The development of new effective antibiotics is a costly endeavor, but to abandon this goal could

lead to severe health problems [6]. Thus, new approaches are urgently needed. Antimicrobial

peptides (AMPs) represent natural products, occurring from microorganisms to the human. AMPs

consist of 15 to nearly 50 amino acids and are generally positively charged [7]. Importantly, many

AMPs are amphiphilic and interact with biomembranes by disrupting the cell membrane. In contrast,

most of the established antibiotics act against targets like ribosomes or enzymes involved in the cell

wall synthesis [8]. Due to their broad-spectrum and bactericidal activities, AMPs are promising

therapeutic alternatives in the fight against multiresistant bacteria [9]. Naturally occurring peptides

are mostly not directly suitable for medical use as therapeutics because they show intrinsic

weaknesses, including poor chemical and physical stability and also a short circulating plasma half-

life [10]. Another drawback of many AMPs is their low efficacy against Gram-negative bacterial

pathogens [7]. Amphibians produce evolutionary optimized AMPS which serve as potent tools

against bacterial infections [11]. Ranalexin, an AMP from the skin of the American bullfrog, Rana

catesbeiana, consists of 20 amino acids (Figure 1) [12]. It is an amphipathic molecule with a cationic

heptapeptide ring at its carboxyl terminus and a hydrophobic region at its amino terminal end [12].

Page 6: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

These two regions are linked via a “hinge” molecule made of the amino acid proline. The cyclic

structure of ranalexin is based on a disulfide bond between the two cysteine residues. Ranalexin

combined with lysostaphin displayed synergistic inhibitory effects against clinical isolates of S. aureus

[13]. Combination of ranalexin with linezolid or polymyxin B displayed significant synergism against

multidrug-resistant bacteria [14]. Ghiselli et al. further tested a single dose treatment of ranalexin in

rats with septic shock, leading to prevention of bacterial growth, endotoxemia and mortality in the

animals [15].

In order to improve the antibacterial activity of ranalexin, modifications of the peptide are needed.

Acylation can significantly improve the antimicrobial potency of peptides [16]. In contrast, the

removal of the acyl chain almost completely destroys the antimicrobial activity of polymyxin B [16].

Due to the fact, that solid-phase peptide synthesis is quite expensive and complex, Aleinein et al.

established a method of expressing recombinant ranalexin in Escherichia coli and Pichia pastoris

which represents an alternative route for peptide production [14, 17].

In this present article, we report on the development of novel lipopeptides based on the naturally

occurring AMP ranalexin. We demonstrate their improved antimicrobial activity against a variety of

Gram-positive and Gram-negative bacteria, including multidrug-resistant clinical isolates. We also

evaluate the substances for their cytotoxicity on various vertebrate cell types. Furthermore, we

delineate their formidable pharmacokinetic profile in a Wistar rat model using positron emission

tomography (PET).

Please insert Figure 1 here

Please insert in Table 1 here

2. Material and methods

2.1 Peptide synthesis

Page 7: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Ranalexin and its lipopeptide derivatives (C14C3lexin, C13C3lexin, C12C3lexin C11C3lexin and

C10C3lexin), were synthesized by fluoren-9-methoxycarbonyl (Fmoc) chemistry with an Applied

Biosystems ABI 433A synthesizer as described in detail by Brings et al. [18]. For further information

see supplementary data.

2.2 Antimicrobial activity

Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens DSM

50090, Salmonella subterranea DSM 16208, Yersinia mollaretii DSM 18520, Bacillus licheniformis

DSM 13, Bacillus megaterium DSM 32, Bacillus subtilis DSM 10 and Staphylococcus auricularis DSM

20609 were purchased from the Leibniz Institute DSMZ German Collection of Microorganisms and

Cell Culture (Braunschweig, Germany). All other bacteria were obtained from the Department of

Medical Microbiology and Hygiene, Heidelberg University Hospital. The MIC values were determined

by broth microdilution method of the Clinical and Laboratory Standards Institute (CLSI, Wayne, PA,

USA) [19]. The protocol is described in detail in the supplementary information.

2.3 Time-kill studies

Time-kill studies were performed according to the guidelines of CLSI [20]. Briefly, Escherichia coli

ATCC 25922 and Staphylococcus aureus ATCC 25923 were adjusted to 1x 108 cfu/ml, diluted to 1x 106

cfu/ml and adjusted to 4x MIC, 2x MIC and 1x MIC of ranalexin, C13C3lexin and cefuroxime (potency

88.6%) (Sigma-Aldrich, Steinheim, Germany) as control. Also, an aliquot of the 1*106 cfu/mL dilution

was mixed to an equal volume of medium (cation-adjusted Mueller Hinton broth) for growth control.

After 0, 0.5, 1, 2, 4, 8 and 12 h aliquots were serially diluted (1:10) in sterile saline. 10 μl of each

dilution were spread onto agar plates. After 24 h of incubation at 37 °C, cfu were counted and the

viable units within the original inoculum were calculated.

2.4 Cell viability assay

Page 8: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

In order to assess cytotoxic effects of the AMPs against eukaryotic tissue, the human tumor cell lines

MCF-7 (breast adenocarcinoma, ACC-115), HeLa (cervical adenocarcinoma, ACC-57), C4-2 (prostate

carcinoma, ATCC CRL-3314), HepG2 (liver hepatocellular carcinoma, ACC-180), the immortalized

Chinese hamster (Cricetulus griseus) ovary cell line CHO-K1 (ACC-110) as well as human peripheral

blood mononuclear cells (PBMCs) were exposed to the peptides. AMP cytotoxicity was investigated

in vitro by an MTT viability assay [21]. The protocol is described in detail in the supplement.

2.5 Radiolabeling with 68Ga

For the PET scans, the DOTA-conjugated peptides were labelled with 68Ga which was eluted with 0.6

M hydrochloric acid (HCl) from an iThemba LABS 68Ge/68Ga generator (DSD Pharma, Pukersdorf,

Austria) as described previously [22] (further information see supplement).

2.6 Micro-PET Imaging in Wistar rats

The procedures in this study were approved by the Animal Care and Use committees at the

Regierungspräsidium Karlsruhe, Germany. Adult, male, Wistar rats (250 – 300 g) were purchased

from Janvier Labs (Le Genest-Saint-Isle, France). The animals were anaesthetized by isoflurane

inhalation and 80 – 100 MBq of the 68Gallium labelled peptide, dissolved in 100 µl of 0.9% NaCl were

injected into the tail vein. Imaging was performed with an Inveon small-animal PET scanner (Siemens,

Erlangen, Germany). Cumulative images from 0 – 20 min, 20 – 40 min and 40 – 60 min after injection

were taken. For the DOTA coupled lipopeptide one further image from 120 – 140 min after injection

was made. The data were normalized for animal weight and injected dose. Pictures and data are

given as standardized uptake value (SUV) [18].

3. Results

3.1 Peptide synthesis

Page 9: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Pure batches of ranalexin and derived lipopeptides C10C3lexin, C11C3lexin, C12C3leixn, C13C3lexin

and C14C3lexin were obtained. Their sequences, molecular masses and the weight analyzed by

HPLC/MS are shown in Table 1. The chemical structures of ranalexin and C13C3lexin are illustrated in

Figure 1, delineating the design of novel lipopeptide based on the conjugation of saturated fatty acid

to a peptide moiety. Detailed LC-MS spectra can be found in supplementary Figures 1 - 4.

3.2 Antimicrobial susceptibility of clinical isolates

The results of the susceptibility testing of the bacterial strain E. faecium UL602570, P. aeruginosa

BL303790 and K. pneumoniae KL800877 with a variety of antibiotics are shown in Table 2. Through

molecular biological analyses diverse resistance genes were detected. E. faecium UL602570 contains

a vanA resistance gene. The metallo-β-lactamase gene blaVIM-2 was found in P. aeruginosa

BL303790. K. pneumoniae KL800877 contains a KPC-2 Klebsiella pneumoniae carbapenemase. The

results of the susceptibility testing of the different clinical Acinetobacter isolates, shown in

supplementary Table 1, were quite alarming. Only A. baumannii BL600773 and A. junii KL511137

showed a wide spectrum of susceptibility to established antibiotics. The other A. baumannii isolates

belong to the group of multiresistant Gram-negative bacteria (MRGN). A. baumannii BL600773;

KL210652; KL314822; SC300007; SR201346; SR300573 and UL515338 are 4-MRGN strains, which are

resistant to antibiotics of the groups of carbapenems, quinolones, amino glycosides and fourth

generation cephalosporins. A. baumannii KL309623; SC322333 and SC404997 belong to the 3-MRGN

strains, being resistant to 3 of the groups mentioned. The antimicrobial susceptibility testing here

confirmed previous findings, that the polymyxin antibiotic colistin is often one of the only few

remaining antibiotics effective against multiresistant A. baumannii strains.

Please insert Table 2 here

3.3 Antimicrobial activity of the AMPs

Page 10: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

The results of the MIC testing of AMPs against a representative selection of bacteria are shown in

Figure 2 and 3. Ranalexin shows a potent activity against all 12 tested Acinetobacter strains (4 – 8

mg/L), and also against Gram-positive bacteria (8 – 16 mg/L). Against the tested Gram-negative non-

Acinetobacter bacteria, ranalexin shows only low antimicrobial activity (16 - >64 mg/L). In contrast,

the lipopeptides derivatives are more potent than ranalexin. Especially, MIC values against the non-

Acinetobacter Gram-negative bacteria are significantly lower for C13C3lexin and C12C3lexin (2 – 16

mg/L) than the MIC values for ranalexin. Tridecanoic acid and dodecanoic acid seem to be the

preferred chain length for the antimicrobial activity. Gram-positive bacteria are less sensitive against

variations in the molecular structure of the antimicrobial peptides (Figure 3). The non-Acinetobacter

Gram-negative bacteria are very sensitive for variations of the chain length of the fatty acid. The

changes of the MIC values demonstrate this fact, especially by comparison up the values for

C11C3lexin (4 – 16 mg/L) and C10C3lexin (16 – 64 mg/L). Another important characteristic of

ranalexin and the lipopeptide derivatives is that they are not affected by resistance mechanisms

against established antibiotics. Even multiresistant strains like K. pneumoniae KL800877 or A.

baumannii SC300007 are sensitive to the lipopeptides. The MICs for the bacteria of the genus

Pseudomonas are of special interest since they are higher than the MICs of the other bacteria tested.

The MBC is mostly one to two potencies higher than the MIC (Table 3). This allows the assumption

that the mechanism of action of the peptides is in fact bactericidal.

Please insert Figure 2 here

Please insert Figure 3 here

Please insert Table 3 here

3.4 Time-kill studies

For estimation of time-kill kinetics, we performed time-kill studies against Gram-negative E. coli ATCC

25922 and Gram-positive S. aureus ATCC 25923 at different concentrations (4x MIC, 2x MIC and 1x

MIC). The results, illustrated in Figure 4, indicate that ranalexin and C13C3lexin are concentration

Page 11: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

dependent and fast acting, bactericidal substances. At a concentration of 4x MIC, no cfu could be

detected after 30 min of incubation. At a concentration of 2x MIC the lipopeptide C13C3lexin shows

an advantage compared to ranalexin. After 30 min no cfu could be detected for the lipopeptide,

whereas for ranalexin, no cfu could be detected only after 2 h for E. coli and after 4 h for S. aureus,

respectively. Both substances, ranalexin and C13C3lexin exhibited superior killing kinetics compared

to the established antibiotic cefuroxime (Figure 4).

Please insert Figure 4 here

3.5 Cell viability assay

Essential for the assessment of any potential clinical application, cytotoxicity of ranalexin and its

derived lipopeptides was investigated in a viability assay against different cancer as well as non-

cancerous cell types. The IC50 values, representing the specific concentrations of the respective

substances tested upon which 50% viability is observed within the chosen cell culture, are displayed

in Figure 5. Our study remarkably shows that the cytotoxicity of the lipopeptides is dependent on the

chain length of the fatty acid linked to the peptide. The AMPs carrying fatty acid moieties from C14

to C12 (IC50 12.74 – 32.07 µg/mL) exhibit higher cytotoxicity than ranalexin (IC50 21.61 – 32.29

µg/mL). In contrast, for C10C3lexin lower toxicity was observed (IC50 38.49 – 108.10 µg/mL). Even

though, the IC50 values differ between the different cell lines, they show a similar trend for ranalexin

as well as for its derivatives. Importantly, also non-tumorous cell lines were tested, such as the

hamster ovary cells CHO and human peripheral blood cells (PBMCs). Even though these cells show

similar sensitivity to the compounds tested as the tumor cells lines, native PBMCs are significantly

less vulnerable to the AMP C10C3lexin (IC50 108.10 µg/mL) carrying the shortest fatty acid moiety.

Please insert Figure 5 here

3.6 Micro PET Imaging

Page 12: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

For estimation of the pharmacokinetics, PET imaging of DOTA-labelled ranalexin and C13C3lexin was

performed in Wistar rats (Figure 6). The corresponding calculated SUV values after injection are

shown in the supplementary Figures 5 and 6. The PET images and the SUVs clearly show that

ranalexin is eliminated through the kidneys and excreted via the urine. Interestingly, the first picture

from 0 – 20 min reveal that a greater amount of radioactivity is localized in the renal pelvis and in the

bladder, followed by renal accumulation. 1 h post administration, most of the radioactivity is found

in the bladder, which indicates a short half-life of ranalexin within the body. In contrast, the

pharmacokinetics of C13C3lexin look very promising. The images demonstrate that the lipopeptide

accumulates within the liver and stays there for several hours. Appreciable amounts of radioactivity

in the bladder are first visible after 40 – 60 min. Most importantly, the modified peptide shows no

accumulation within the kidneys. The images strongly indicate a prolonged retention time of

C13C3lexin within the animal body. This is exemplarily shown by determination of the SUV of the

heart for both peptides over a period of 60 min post injection in Figure 7. The SUV, the area under

the curve (AUC) and the plasma half-life are significantly higher for the lipopeptide than for the

natural peptide. These results clearly indicate the remarkably improved pharmacokinetics of DOTA-

C13C3lexin compared to DOTA-ranalexin.

Please insert Figure 6 here

Please insert Figure 7 here

4. Discussion

The number of new antibiotic drugs currently in clinical phase 2 or 3 testings still remains sobering.

The lack of new antibiotics against multidrug resistant Gram-negative bacteria endanger patients [2].

Especially the antibiograms of clinical isolates (Table 2) illustrate this problem. The emergence of

MRGN bacteria and the lack of new antibiotics with an innovative mode of action caused a revival of

polymyxins [23]. Polymyxins are considered one of the few remaining last-line antibiotics against

infections with drug-resistant Gram-negative pathogens [24]. The antimicrobial susceptibility testing

Page 13: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

here confirmed previous findings, that the polymyxin antibiotic colistin is often one of the only few

remaining antibiotics effective against multiresistant A. baumannii strains. Most of multidrug

resistant infections, from intensive care units, are caused by A. baumannii and K. pneumoniae [25].

Thus, we included multidrug-resistant isolates of these species in our study. Besides these well-

known problems, multiresistant Gram-positive bacteria also occur more frequently. Although most of

the multiresistant Gram-positive infections can still be treated with vancomycin, it is necessary to

develop new, innovative antibiotics to counterattack currently increasing antimicrobial resistances in

Enterococci and Staphylococci [26].

In this study, we investigated the antimicrobial activity of ranalexin and its lipopeptide derivatives

C10C3lexin, C11C3lexin, C12C3lexin, C13C3lexin and C14C3lexin. The peptides were synthesized with

solid-phase peptide synthesis by fmoc-chemistry. Solid-phase peptide synthesis represents an

effective and fast method of for peptide production [27]. For ranalexin we confirmed a high activity

against Gram-positive bacteria but weaker activity against Gram-negative bacteria. The MIC values

were comparable with those found in previous publications [12]. For E. faecalis ATCC 29212, P.

aeruginosa ATCC 27853 and E. coli ATCC 25922 quality control ranges of MICs for vancomycin and

colistin, respectively are provided by the CLSI [19]. The MIC values of the positive controls in our

study are in accordance with those quality control MICs, which indicates the correct performance of

the broth dilution method. The approved lipopeptide antibiotics colistin and daptomycin possess N-

terminal fatty acids which are essential for their antimicrobial activity [28]. N-acylation of AMPs could

lead to enhanced antimicrobial activity [29]. In this study, we successfully demonstrated, that

shortened, acylated derivatives of ranalexin are significantly more potent against Gram-positive and

Gram-negative bacteria, compared with the naturally occurring peptide. The remarkably broad

spectrum of the antimicrobial activity of the lipopeptides indicates their potential as novel highly

effective antibiotics. A broad spectrum antibiotic could be especially used for the treatment of

bacterial infections with any unknown pathogenic strain [30].

We also demonstrated that acylation is a powerful tool in order to improve the activity against Gram-

negative bacteria but this class of bacteria is also very sensitive to changes in the length of the fatty

Page 14: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

acid. Even small variations had significant influence on antimicrobial activity. As a fact, Gram-

negative bacteria contain a thinner cell wall peptidoglycan layer but possess an additional outer

membrane which functions as a permeability barrier [31]. For this reason, it is more difficult for

antibiotics to reach the inner cell membrane. Acylation could facilitate the penetration through the

outer membrane [8]. Indeed, the synthesized lipopeptides carry the same amino acid sequence. The

only difference is the length of the fatty acid attached. In the cut-out of the HPLC profile of ranalexin

and the lipopeptides (Supplementary Figure 7) it is clearly visible that an increase in length of the

fatty acids causes a prolongation of the retention time. It is well known, that the retention time is

proportional to the hydrophobicity of the respective peptides [32]. Thus, C12C3lexin and C13C3lexin

probably possess the ideal hydrophobicity for penetrating the outer membrane of Gram-negative

bacteria. Furthermore, the peptides show a bactericidal mode of action and most importantly, AMPs

are not affected by classical mechanisms of resistance which has evolved against antibiotics attacking

differing targets, such as ribosomes, the cell wall or DNA [33]. The antimicrobial activity of ranalexin

and the lipopeptides are moderate against P. fluorescens DSM 50090, compared to other Gram-

negative bacteria. As a possible explanation, it can be assumed that P. fluorescens belongs to the

resident skin flora of R. catesbeiana and therefore also produces the antibiotic mupirocin [34, 35].

Time-kill studies confirmed the speed of bactericidal activity as another powerful advantage of the

peptides [36]. Established bactericidal antibiotics like cefuroxime need several hours for killing

infectious bacteria [37], whereas ranalexin and C13C3lexin showed improved bactericidal kinetics.

The superior bactericidal kinetics of C13C3lexin clearly highlights the potential of lipopeptides as fast

acting bactericidal substances. Because of their membrane-targeted mode of action, AMPs obviously

bear a potential for toxicity [38]. With respect to future clinical application, a fond estimation of

cytotoxicity of ranalexin and the lipopeptide derivatives against vertebrate cells is essential. Using a

cell viability assay, we were able to assess their cytotoxicity against several cell lines, and most

importantly, also against non-tumorous hamster and native human blood cells. While ranalexin

showed moderate toxicity (IC50 21.62 – 32.29 µg/mL) compared to other AMPs [39], the harmful toxic

activity of the lipopeptides obviously depends on the chain length of the fatty acid attached to the

Page 15: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

peptidic moiety. Fatty acids comprising chain lengths from C14 to C12 showed unfavorable toxicity

(IC50 12.74 – 32.07 µg/mL), whereas a chain length of C10, represented by decanoic acid, significantly

reduced the toxicity of the lipopeptide (IC50 38.49 – 108.10 µg/mL). Interestingly, human PBMCs

were shown to exhibit a dramatically lower sensitivity towards the C10C3lexin compound compared

to all other cell types tested. Although, in general, the influence of the fatty acid on toxicity is still

moderate compared to the influence on antimicrobial activity, our observations necessitate further

investigations to clearly reveal the impact of specific modifications of AMPs on both improved

antimicrobial activity and the reduction of potential adverse effects.

It was possible for the first time to estimate the pharmacokinetics of ranalexin in rats. The

biodistribution studies, performed with PET-imaging of DOTA-ranalexin and DOTA-C13C3lexin in

Wistar rats showed promising improvements with respect to pharmacokinetics. While the natural

peptide ranalexin is rapidly cleared via the kidneys and excreted through the urine, the synthetic

lipopeptide accumulates in the liver for a minimum of two hours and shows prolonged circulation

within the blood. Acylation has been proven to be a reliable tool for improving the half-life of a drug.

Telavancin, a lipoderivative of vancomycin, which was approved in 2009, has a half-life in humans of

9 h compared to 6 h of its origin vancomycin [30, 40]. The lately approved dalbavancin, another

lipoderivative of vancomycin, shows a half-life of over 300 h in humans [30]. Short half-lives of

antimicrobial peptides is a serious and well-known problem [38]. Therefore, we could show that the

modification of the antimicrobial peptide ranalexin by the replacement of particular amino acids with

tridecanoic acid leads to improved antimicrobial activity, prolonged half-life as well as specific

targeting into the liver. The SUV of the heart of a radiolabeled compound is a good indicator for the

residence time of a substance within the blood stream. The comparison of the SUVs of the heart of

DOTA-ranalexin and DOTA-C13C3lexin clearly evidences that the lipopeptide shows a longer plasma

half-life. Furthermore, the higher SUVs of the lipopeptide clearly demonstrate that the lipopeptide

shows superior pharmacokinetic characteristics compared with the natural peptide.

The lipopeptide derivatives studied contain only 14 amino acids instead of 20 amino acids of the

origin ranalexin. The costs for synthesizing the peptides are 5 to 20 times higher than for classical

Page 16: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

antibiotics produced by fermentation [33]. Therefore, modification of ranalexin by lipopeptides

shows another big advantage, because the production costs could be reduced. In conclusion, our

investigation shows that the examined lipopeptides represent one promising example that new

substances can be generated from evolutionary optimized natural products using the knowledge of

modern drug design.

The enhanced antimicrobial activity and the improved pharmacokinetics of the lipopeptide

derivatives of ranalexin could lead to promising new drug candidates for the management of

occurring microbial challenges. Nevertheless, the described novel class of lipopeptides is by no

means the end of the story. Further improvements are needed for the design of a clinically applicable

lipopeptidic AMP in order to find the optimal balance between effective antimicrobial activity on the

one hand and minimal detrimental impact on the treated patient on the other. Moreover, since the

main part of the lipopeptides comprises an amino acid backbone which is naturally prone to

degradation by proteases in the gut, the issue of oral bioavailability will also be an important task for

future developments of AMP therapeutics.

Declarations

Funding: No funding

Competing Interests: No conflict of interest

Ethical Approval: Not required

Page 17: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

References

[1] Ling LL, Schneider T, Peoples AJ, Spoering AL, Engels I, Conlon BP, et al. A new antibiotic kills pathogens without detectable resistance. Nature. 2015;517:455-9. [2] Boucher HW, Talbot GH, Bradley JS, Edwards JE, Gilbert D, Rice LB, et al. Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of America. Clin Infect Dis. 2009;48:1-12. [3] Centers for Disease Control and Prevention, Antibiotic Resistance Threats in the United States, https://www.cdc.gov/drugresistance/threat-report-2013/index.html; 2013 [accessed 17.10.23]. [4] Rice LB. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J Infect Dis. 2008;197:1079-81. [5] Maragakis LL, Perl TM. Acinetobacter baumannii: epidemiology, antimicrobial resistance, and treatment options. Clin Infect Dis. 2008;46:1254-63. [6] Hancock REW. The end of an era? Nature Reviews Drug Discovery. 2007;6:28-. [7] Fox JL. Antimicrobial peptides stage a comeback. Nat Biotechnol. 2013;31:379-82. [8] Makovitzki A, Avrahami D, Shai Y. Ultrashort antibacterial and antifungal lipopeptides. Proc Natl Acad Sci U S A. 2006;103:15997-6002. [9] Kanthawong S, Nazmi K, Wongratanacheewin S, Bolscher JG, Wuthiekanun V, Taweechaisupapong S. In vitro susceptibility of Burkholderia pseudomallei to antimicrobial peptides. Int J Antimicrob Agents. 2009;34:309-14. [10] Fosgerau K, Hoffmann T. Peptide therapeutics: current status and future directions. Drug Discov Today. 2015;20:122-8. [11] Simmaco M, Mignogna G, Barra D. Antimicrobial peptides from amphibian skin: what do they tell us? Biopolymers. 1998;47:435-50. [12] Clark DP, Durell S, Maloy WL, Zasloff M. Ranalexin. A novel antimicrobial peptide from bullfrog (Rana catesbeiana) skin, structurally related to the bacterial antibiotic, polymyxin. Journal of Biological Chemistry. 1994;269:10849-55. [13] Graham S, Coote PJ. Potent, synergistic inhibition of Staphylococcus aureus upon exposure to a combination of the endopeptidase lysostaphin and the cationic peptide ranalexin. J Antimicrob Chemother. 2007;59:759-62. [14] Aleinein RA, Schafer H, Wink M. Secretory ranalexin produced in recombinant Pichia pastoris exhibits additive or synergistic bactericidal activity when used in combination with polymyxin B or linezolid against multi-drug resistant bacteria. Biotechnol J. 2014;9:110-9. [15] Ghiselli R, Giacometti A, Cirioni O, Orlando F, Mocchegiani F, Pacci AM, et al. Therapeutic efficacy of the polymyxin-like peptide ranalexin in an experimental model of endotoxemia. J Surg Res. 2001;100:183-8. [16] Jerala R. Synthetic lipopeptides: a novel class of anti-infectives. Expert Opin Investig Drugs. 2007;16:1159-69. [17] Aleinein RA, Hamoud R, Schafer H, Wink M. Molecular cloning and expression of ranalexin, a bioactive antimicrobial peptide from Rana catesbeiana in Escherichia coli and assessments of its biological activities. Appl Microbiol Biotechnol. 2013;97:3535-43. [18] Brings S, Fleming T, De Buhr S, Beijer B, Lindner T, Wischnjow A, et al. A scavenger peptide prevents methylglyoxal induced pain in mice. Biochim Biophys Acta. 2016;1863:654-62. [19] CLSI. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically; approved standard—Ninth edition. CLSI document M07-A9. Wayne, PA: Clinical and Laboratory Standards Institute; 2012. [20] NCCLS. Methods for determining bactericidal activity of antimicrobial agents; Approved guideline. NCCLS document M26-A [ISBN 1-56238-384-1]. NCCLS, 940 West Valley Road, Suite 1400, Wayne, Pennsylvania 19087 USA, 1999. [21] Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65:55-63. [22] Wischnjow A, Sarko D, Janzer M, Kaufman C, Beijer B, Brings S, et al. Renal Targeting: Peptide-Based Drug Delivery to Proximal Tubule Cells. Bioconjug Chem. 2016;27:1050-7.

Page 18: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

[23] Falagas ME, Kasiakou SK. Colistin: the revival of polymyxins for the management of multidrug-resistant gram-negative bacterial infections. Clin Infect Dis. 2005;40:1333-41. [24] Ortiz de la Tabla V, Ortega A, Bunuel F, Perez-Vazquez M, Marcos B, Oteo J. Detection of the high-risk clone ST131 of Escherichia coli carrying the colistin resistance gene mcr-1 and causing acute peritonitis. Int J Antimicrob Agents. 2017;49:115-6. [25] Mezzatesta ML, Caio C, Gona F, Cormaci R, Salerno I, Zingali T, et al. Carbapenem and multidrug resistance in Gram-negative bacteria in a single centre in Italy: considerations on in vitro assay of active drugs. Int J Antimicrob Agents. 2014;44:112-6. [26] Biedenbach DJ, Arhin FF, Moeck G, Lynch TF, Sahm DF. In vitro activity of oritavancin and comparator agents against staphylococci, streptococci and enterococci from clinical infections in Europe and North America, 2011-2014. Int J Antimicrob Agents. 2015;46:674-81. [27] Made V, Els-Heindl S, Beck-Sickinger AG. Automated solid-phase peptide synthesis to obtain therapeutic peptides. Beilstein J Org Chem. 2014;10:1197-212. [28] Hu Y, Amin MN, Padhee S, Wang RE, Qiao Q, Bai G, et al. Lipidated peptidomimetics with improved antimicrobial activity. ACS Med Chem Lett. 2012;3:683-6. [29] Mak P, Pohl J, Dubin A, Reed MS, Bowers SE, Fallon MT, et al. The increased bactericidal activity of a fatty acid-modified synthetic antimicrobial peptide of human cathepsin G correlates with its enhanced capacity to interact with model membranes. Int J Antimicrob Agents. 2003;21:13-9. [30] Singh SB, Young K, Silver LL. What is an "ideal" antibiotic? Discovery challenges and path forward. Biochem Pharmacol. 2017. [31] Hancock RE, Rozek A. Role of membranes in the activities of antimicrobial cationic peptides. FEMS Microbiol Lett. 2002;206:143-9. [32] Yamagami C, Kawase K, Iwaki K. Hydrophobicity parameters determined by reversed-phase liquid chromatography. XV: optimal conditions for prediction of log P(oct) by using RP-HPLC procedures. Chem Pharm Bull (Tokyo). 2002;50:1578-83. [33] Andrea Giuliani GP, Silvia Fabiole Nicoletto. Antimicrobial peptides: an overview of a promising class of therapeutics. CENT EUR J BIOL. 2007;2:1 - 33. [34] Lauer A, Hernandez T. Cutaneous bacterial species from Lithobates catesbeianus can inhibit pathogenic dermatophytes. Mycopathologia. 2015;179:259-68. [35] Fuller AT, Mellows G, Woolford M, Banks GT, Barrow KD, Chain EB. Pseudomonic acid: an antibiotic produced by Pseudomonas fluorescens. Nature. 1971;234:416-7. [36] Mohamed MF, Hammac GK, Guptill L, Seleem MN. Antibacterial activity of novel cationic peptides against clinical isolates of multi-drug resistant Staphylococcus pseudintermedius from infected dogs. PLoS One. 2014;9:e116259. [37] Nielsen EI, Viberg A, Lowdin E, Cars O, Karlsson MO, Sandstrom M. Semimechanistic pharmacokinetic/pharmacodynamic model for assessment of activity of antibacterial agents from time-kill curve experiments. Antimicrob Agents Chemother. 2007;51:128-36. [38] Hancock RE, Sahl HG. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat Biotechnol. 2006;24:1551-7. [39] Vaucher RA, da Motta Ade S, Brandelli A. Evaluation of the in vitro cytotoxicity of the antimicrobial peptide P34. Cell Biol Int. 2010;34:317-23. [40] Rotschafer JC, Crossley K, Zaske DE, Mead K, Sawchuk RJ, Solem LD. Pharmacokinetics of vancomycin: observations in 28 patients and dosage recommendations. Antimicrob Agents Chemother. 1982;22:391-4.

Page 19: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Figure 1: Structure of ranalexin and C13C3lexin. Six N-terminal amino acids are substituted by tridecanoic acid

Page 20: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Figure 2: (A) MICs of ranalexin and lipopeptides against a representative selection of Gram negative bacteria, including multiresistant clinical isolates. Lipopeptides, especially the most potent lipopeptide C13C3lexin, show greatly enhanced antimicrobial activity. A special case are the MICs against Acinetobacter. Ranalexin has potent antimicrobial activity against Acinetobacter, the lipopeptides show comparable MICs, independently from the chain length of the fatty acid. (B) MICs of ranalexin and lipopeptides against a selection of Pseudomonas strains. While the peptides show strong activity against the reference strain P. aeruginosa ATCC 27853, they exhibit reduced activity against P. aeruginosa BL303790* VIM-2 and P. fluorescens DSM 50090. * = multiresistant clinical isolates.

Page 21: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Figure 3: Minimum inhibitory concentration (MIC) of ranalexin and lipopeptides against a

representative selection of Gram positive bacteria. The most potent lipopeptides C13C3lexin

and C12C3lexin show enhanced antimicrobial activity compared to ranalexin. *

multiresistant clinical isolate.

Page 22: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Figure 4: Time-kill curves of ranalexin, C13C3lexin and cefuroxime at 4x MIC, 2x MIC and 1x MIC against S. aureus ATCC 25923 and E. coli ATCC 25922. The peptides are fast acting bactericidal substances. At a concentration of 4x MIC no living bacteria could be detected after 30 min of incubation. Apparently, the lipopeptide C13C3lexin has an enhanced bactericidal activity compared to ranalexin.

E. coli ATCC 25922 S. aureus ATCC 25923

Time-kill studies

Figure 4: Time-kill curves of ranalexin, C13C3lexin and cefuroxime at 4x MIC, 2x MIC and 1x MIC against S. aureus ATCC 25923 and E. coli ATCC 25922. The peptides are fast acting bactericidal substances. At a concentration of 4x MIC no living bacteria could be detected after 30 min of incubation. Apparently, the lipopeptide C13C3lexin has an enhanced bactericidal activity compared to ranalexin.

Page 23: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Figure 5: Cytotoxic capacity of the AMP ranalexin and its derivatives against human cells. Several tumor cells lines of different origin (breast – MCF-7, liver – HepG2, cervix – HeLa, prostate – C4-2), the immortalized Chinese hamster ovary cell line CHO and freshly isolated human peripheral blood mononuclear cells (PBMCs) were cultured upon exposure to increasing concentrations (0.25 - 128 µg/mL) of ranalexin and its derived lipopeptides C10C3lexin, C11C3lexin, C12C3lexin, C13C3lexin and C14C3lexin, carrying fatty acids of various chain lengths (from C10 to C14). The concentrations of the respective AMPs at which 50% cell viability (IC

50) was still observed for the studied cell types, were

estimated by a colorimetric MTT-assay. Cytotoxicity of the lipopeptides depends on the length of the conjugated fatty acid. Lipopeptides containing fatty acids of short chain length, such as C10 and C11, show lower toxicity in comparison to the natural peptide.

Page 24: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Figure 6: PET-images of DOTA-ranalexin (A) and DOTA-C13C3lexin (B) after intravenous injection into the tail vein. After one hour the major part of DOTA-ranalexin is found in the bladder and the renal pelvis. In contrast, the biodistribution of DOTA-C13C3lexin. After two hours of injection, a larger amount of the substance accumulates in the liver.

Page 25: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Figure 7: Standard uptake volume (SUV) of DOTA-C13C3lexin and DOTA-ranalexin in the heart. The systemic availability of the lipopeptide is distinctly improved.

Page 26: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Table 1: Amino acid sequences. theoretical mass [Da] and molecular weight [m/z] after LC-MS

analysis of the investigated peptides. 1

position of DOTA-coupling. 2

LC-MS spectra shown in supplementary data.

peptide sequence theoretical mass

[Da] m/z = 1 m/z = 2 m/z = 3 ranalexin

2

FLGGLIKIVPAMICAVTKKC 2013.1890 2014.1183 1052.5655 702.0463

C14C3lexin myristic acid -

KIVPAMICAVTKKC 1713.0239 1713.9708 856.9888 571.6620

C13C3lexin tridecanoic acid -

KIVPAMICAVTKKC 1699.0082 1698.9459 849.9778

C12C3lexin dodecanoic acid -

KIVPAMICAVTKKC 1684.9926 1684.9316 842.9707 -

C11C3lexin2

undecanoic acid -

KIVPAMICAVTKKC 1670.9769 1670.9769 835.9921 557.6638

C10C3lexin decanoic acid -

KIVPAMICAVTKKC 1656.9613 1656.9080 828.9588 DOTA-

ranalexin2 FLGGLIKIVPAMICAVTKK

1C

2489.3692 - 1245.6499 830.7695 DOTA-

C13C3lexin2

tridecanoic acid -

KIVPAMICAVTKK1C 2085.1884 - 1043.5614 696.0438

Page 27: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Table 2: Susceptibility of the multiresistant isolates to common antibiotics. The isolates are resistant (R) to a multitude of common antibiotics. Only a few antibiotics remain susceptible (S) against these isolates. E. faecium is vancomycin-resistant (VRE). The isolate of P. aeruginosa is a producer of the metallo-β-lactamase VIM-2. K. pneumoniae is a producer of an K. pneumoniae carbapenemase (KPC-2). A. baumannii isolates are resistant to four major groups of antibiotics (4-MRGN). i = intermediate, nt = not tested

Enterococcus faecium UL602570

Pseudomonas aeruginosa BL303790

Klebsiella pneumoniae KL800877

Acinetobacter baumannii KL314822

Acinetobacter baumannii SC300007

VRE VIM-2 KPC-2 4-MRGN 4-MRGN Ampicillin R nt R nt nt Amoxycillin/ Clavulanate

R R R R nt Piperacillin nt R R nt R Piperacillin/ Tazobactam

nt R R R R Cefazoline R nt R nt nt Cefotaxime nt R R R R Ceftriaxone nt R nt R R Ceftazidime nt R R R R Imipenem nt R R R R Meropenem R R R R R Ciprofloxacin R R R R R Gentamicin R S I R R Tobramycin nt R R R R Tigecycline nt R I nt nt Trimethoprim/ Sulfamethoxazole

R R R S R Tetracycline S nt I R nt Vancomycin R nt nt nt nt Teicoplanin R nt nt nt nt Erythromycin R nt nt nt nt Linezolid S nt nt nt nt Colistin nt nt S nt S

Page 28: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

Table 3: Comparison of minimum inhibitory concentration (MIC) and MBC (minimum bactericidal concentration) against a representative variety of Gram-negative and Gram-positive bacteria including multidrug-resistant strains. MIC and MBC in [mg/L]. Obviously, MBC is mostly equal to MIC or only one potency higher, which indicates a bactericidal mode of action of the peptides. An

exception are MBC values of Pseudomonas, that are significantly higher than MIC values. 1

clinical

isolate, 2multiple drug resistance,

3 multiresistant Gram-negative bacteria, resistant to four different

groups of antibiotics, 4

enterohemorrhagic E. coli, 5

K. pneumoniae carbapenemase, 6

Verona

integron-encoded metallo-β-lactamase, 7

vancomycin-resistant Enterococcus, n.i. = no inhibitory activity at 64 mg/L, n.b. = no bactericidal activity at 64 mg/L.

ranalexin

C14C3lexin

C13C3lexin

C12C3lexin

C11C3lexin

C10C3lexin

MIC MBC MIC MBC MIC

MBC MIC MBC MIC MBC MIC MBC Gram-negative bacteria A. baumannii KL314822

1

MDR2

4-MRGN3 4 4 4 4 4 4 2 4 4 4 4 4

A. baumannii SC3000071

MDR2

4-MRGN3 4 8 4 8 2 2 4 4 4 4 4 4

E. coli ATCC 25922 16 32 8 8 4 8 4 4 4 8 8 8 E. coli O157:H7 ATCC 35150

EHEC4 32 32 8 16 4 8 4 8 4 16 8 32

K. pneumoniae KL8008771

MDR2 KPC

5 n.i. n.i. 16 32 8 16 8 8 8 16 32 64 P. aeruginosa BL303790

1

MDR2 VIM-2

6 n.i. n.i. 16 64 8 64 16 64 16 64 32 64 S. enterica DSM 554 64 n.b. 16 32 8 32 16 16 8 8 16 16 Y. mollaretii DSM 18520 n.i. n.i. 4 8 4 8 8 32 8 32 64 64 Gram-positive bacteria B. megaterium DSM 32 8 8 4 4 2 2 4 4 4 4 8 8 B. subtilis DSM 10 8 8 8 8 4 4 4 4 4 4 4 4 E. faecalis DSM 29212 16 16 8 8 4 4 4 4 8 8 8 16 E. faecium UL602570

1

MDR2

VRE7 16 16 4 8 4 4 4 4 4 8 16 16

S. aureus MRSA NCTC

10442 8 16 4 8 4 8 4 4 8 8 16 16 S. epidermidis ATCC 14990 16 16 16 16 8 8 4 4 4 8 8 8

Page 29: A novel tool against multiresistant bacterial pathogens ...€¦ · 2.2 Antimic robial activity Acinetobacter bohemicus DSM 100419, Escherichia coli K12 DSM 498, Pseudomonas fluorescens

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIP

T

GRAPHICAL ABSTRACT